MX2013000395A - Methods of generating natural killer cells. - Google Patents

Methods of generating natural killer cells.

Info

Publication number
MX2013000395A
MX2013000395A MX2013000395A MX2013000395A MX2013000395A MX 2013000395 A MX2013000395 A MX 2013000395A MX 2013000395 A MX2013000395 A MX 2013000395A MX 2013000395 A MX2013000395 A MX 2013000395A MX 2013000395 A MX2013000395 A MX 2013000395A
Authority
MX
Mexico
Prior art keywords
cells
medium
cell
hematopoietic
placental
Prior art date
Application number
MX2013000395A
Other languages
Spanish (es)
Other versions
MX342995B (en
Inventor
Robert J Hariri
Andrew Zeitlin
Mohammad A Heidaran
Xiaokui Zhang
Ajai Pal
Eric Law
Vanessa Voskinarian-Berse
Lin Kang
Stephen Jasko
Bhavani Stout
Original Assignee
Anthrogenesis Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Anthrogenesis Corp filed Critical Anthrogenesis Corp
Publication of MX2013000395A publication Critical patent/MX2013000395A/en
Publication of MX342995B publication Critical patent/MX342995B/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464499Undefined tumor antigens, e.g. tumor lysate or antigens targeted by cells isolated from tumor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • C12N2500/25Insulin-transferrin; Insulin-transferrin-selenium
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/32Amino acids
    • C12N2500/33Amino acids other than alpha-amino carboxylic acids, e.g. beta-amino acids, taurine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/36Lipids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/38Vitamins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/44Thiols, e.g. mercaptoethanol
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/46Amines, e.g. putrescine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/70Undefined extracts
    • C12N2500/80Undefined extracts from animals
    • C12N2500/84Undefined extracts from animals from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/125Stem cell factor [SCF], c-kit ligand [KL]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/145Thrombopoietin [TPO]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2302Interleukin-2 (IL-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2303Interleukin-3 (IL-3)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2307Interleukin-7 (IL-7)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2315Interleukin-15 (IL-15)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/26Flt-3 ligand (CD135L, flk-2 ligand)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/59Lectins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/90Polysaccharides
    • C12N2501/91Heparin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/998Proteins not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/11Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from blood or immune system cells
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

Provided herein are methods of producing natural killer cells using a two-step expansion and differentiation method. Also provided herein are methods of suppressing tumor cell proliferation, of treating individuals having cancer or a viral infection, comprising administering the NK cells produced by the method to an individual having the cancer or viral infection.

Description

METHODS OF GENERATING NATURAL KILLING CELLS 1. RELATED REQUESTS This application claims the benefit of the provisional patent application of E.U.A. Serial No. 61 / 363,981, filed July 13, 2010 and provisional patent application of E.U.A. Serial No. 61 / 497,897, filed on June 16, 2011, the descriptions of which are hereby incorporated by reference in their entirety. 2. FIELD OF THE INVENTION Methods of producing a population of natural killer cells, eg, natural killer cells derived from placenta, eg, placental perfusate (eg, perfused human placenta) such as natural killer cells derived from placenta, are provided herein. , or other tissues, for example, umbilical cord blood or peripheral blood. Also present are populations of expanded natural killer cells produced by the methods presented herein. In addition, methods of using placental perfusate, and natural killer cells thereof, to suppress the proliferation of tumor cells are provided herein. In certain embodiments, natural killer cells are used in combination with, or treated with, one or more immunomodulatory compounds, for example, immunomodulatory compounds referred to as IMiDs ™. 3. BACKGROUND OF THE INVENTION Natural killer (NK) cells are cytotoxic lymphocytes that are a major component of the innate immune system. NK cells do not express receptors for T-cell antigen (TCR), CD3 or B-cell surface immunoglobulin receptor (Ig). NK cells usually express the surface markers CD16 (FcyRIII) and CD56 in humans, but a subclass of human NK cells is CD16. NK cells are cytotoxic; Small granules in your cytoplasm contain special proteins such as perforin and proteases known as granzymes. When released in close proximity to a cell targeted for killing, perforin forms pores in the cell membrane of the target cell through which granzymes and associated molecules can enter, inducing apoptosis. A granzyme, granzyme B (also known as granzyme 2 and serine esterase 1 associated with cytotoxic T lymphocyte), is a serine protease crucial for rapid induction of target cell apoptosis in the cell-mediated immune response.
NK cells are activated in response to interferons or cytokines derived from macrophages. Activated NK cells are referred to as killer cells activated with lymphokine (LAK). NK cells possess two types of surface receptors, labeled "activating receptors" and "inhibitory receptors" that control the cytotoxic activity of cells.
Among other activities, NK cells play a role in tumor host rejection. Since cancer cells have reduced expression or none of MHC class I, they become targeted by NK cells. Accumulating clinical data suggest that haploidentic transplantation of human NK cells isolated from peripheral blood mononuclear cells (PBMC) or potent antileukemia effects mediated bone marrow without detectable graft that incurs host disease (GVHD). See uggeri et al., Science 295: 2097-2100 (2002). Natural killer cells can be activated by cells lacking, or exhibiting reduced levels of, major histocompatibility complex (MHC) proteins. Additionally, activating receptors expressed in NK cells are known to mediate the detection of cells "stressed" or transformed with express ligands to activating receptors and thus trigger NK cell activation. For example, NCR1 (NKp46) binds viral hemagglutinins. Ligands of NKG2D include CMV binding protein 1 UL16 (ULB1), ULB2, ULB3 and sequence A related to MHC class I polypeptide (MICA) and MICB proteins. The protein NK 2B4 binds CD48, and DNAM- a poliovirus receptor (PVR) and Nectin-2, both are consistently connected in acute myeloid leukemia (AML). See Penda et al., Blood 105: 2066-2073 (2004). In addition, AML lysis has been described primarily as a natural cytotoxicity (NCR) receptor dependent. See Fauriat et al., Blood 109: 323-330 (2007). Activated and expanded NK cells and peripheral blood LAK cells have been used in ex vivo therapy and in vivo treatment of patients who have advanced cancer, with some success against bone marrow related diseases, such as leukemia.; breast cancer; and certain types of lymphoma. LAK cell treatment requires that the patient first receive IL-2, followed by leukopheresis and then an ex vivo incubation and culture of autologous lymphocytes harvested in the presence of IL-2 for a few days. LAK cells are due to complete therapy. This purging treatment is expensive and can cause serious side effects. These include fluid retention, pulmonary edema, drop in blood pressure, and high fever.
Despite the advantageous properties of NK cells in killer tumor cells and virus infected cells, they remain difficult to work with and apply in immunotherapy, primarily because of the difficulty of maintaining their tumor and tumoricidal targeting abilities during culture and expression. In this way, it is necessary in the art to develop an efficient method to produce and expand natural killer cells that retain tumoricidal functions.
BRIEF DESCRIPTION OF THE INVENTION In the present methods are provided for expanding and differentiating cells, for example, hematopoietic cells, such as hematopoietic stem cells, for example, CD34 + hematopoietic stem cells, to natural killer cells. In one aspect, a method of producing natural killer (NK) cells comprising culturing hematopoietic stem cells or progenitor cells, e.g., stem cells or CD34 + progenitor cells, is provided in a first medium to produce expanded and differentiated cells, and subsequently cultivating said expanded cells in a second medium wherein said cells expand further and differentiate into natural killer cells. The first and second steps involve cultivating the cells in medium with a unique combination of cellular factors. In certain embodiments, said cellular factors (e.g., cytokines) are not comprised within an undefined component of the medium (e.g., serum), e.g., cellular factors (e.g., cytokines) are exogenous to the undefined component of the medium (for example, serum). In certain modalities, said method is a two-step method. In certain embodiments, said method does not comprise any third step or intermediate where the cells are contacted. In a specific embodiment, a method of producing a population of activated natural killer (NK) cells is provided herein, comprising: (a) planting a population of stem cells or hematopoietic progenitors in a first medium comprising interleukin-15 (IL-15) and, optionally, one or more of the stem cell factor (SCF) and interleukin-7 (IL-7), wherein said optional IL-15 and SCF and IL-7 are not comprised within an undefined component of said medium, so that the population expands, and a plurality of hematopoietic stem or progenitor cells within said population of hematopoietic stem or progenitor cells differentiate into NK cells during said expansion; and (b) expanding the cells of the first passage in a second medium comprising interleukin-2 (IL-2), to produce a population of activated NK cells. Natural killer cells produced by the methods provided herein (eg, two-step method) are referred to herein as TSNK cells.
In certain embodiments, said first medium comprises medium comprising one or more of human serum (e.g., human serum AB), fetal bovine serum (FBS) or fetal calf serum (FCS), e.g., 5% to 20% v / v, stem cell factor (SCF), for example, 1 ng / mL at 50 ng / mL, tyrosine kinase-3 ligand of FMS type (ligand Flt-3), for example, 1 ng / ml at 20 ng / mL; interleukin-7 (IL-7), for example, 1 ng / mL at 50 ng / mL; thrombopoietin (TPO), for example, 1 ng / mL at 50 ng / mL; Interleukin-2 (IL-2), for example, 50 IU / mL at 500 IU / mL; interleukin-15 (IL-5), for example, 1 ng / mL at 50 ng / mL; and / or heparin, for example, 0.1 lU / mL at 10 lU / mL. In a specific embodiment, said first medium comprises the stem cell factor (SCF), interleukin-7 (I L-7) and interleukin-15 (IL-15). In another specific embodiment, said first medium comprises growth medium, human serum (e.g., human serum AB), FBS FCS, SCF, IL-7 and IL-15. In another specific embodiment, said first medium further comprises ligand Flt-3 (Flt3-L), IL-2 and / or heparin. In another specific embodiment, said first medium comprises growth medium, 10% human serum or fetal bovine serum., 20 ng / mL SCF, 10 ng / ml Flt3-L, 20 ng / mL IL-7, 20 ng / mL TPO, 200 lU / mL IL-2, 10 ng / mL IL-15, and 1.5 lU / mL heparin. In another specific embodiment, said first medium does not comprise IL-2. In another specific embodiment, said culture in said first medium comprises culture using feeder cells, e.g., K562 cells, e.g., K562 cells treated with mitomycin C, peripheral blood mononuclear cells (PBMCs), e.g., PBMCs treated with mitomycin C or tissue culture adherent stem cells, e.g., tissue culture adherent stem cells treated with mitomycin C.
In certain embodiments, said first medium is, or comprises, GBGM®, AIM-V®, X-VIVO ™ 10, X-VIVO ™ 15, OPTIMIZER, STEMSPAN® H3000, CELLGRO COMPLETE ™, and / or DMEM.F12. In certain embodiments, said medium comprises O-acetyl-carnitine (also referred to as acetylcarnitine, O-acetyl-L-carnitine or OAC), for example, about 0.5 mM-10 mM. In one embodiment, said medium comprises Stemspan® H3000 and / or DMEM: F12 and OAC, for example, about 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mM. In a specific embodiment, said means comprises GBGM®. In another specific embodiment, said medium comprises DMEM.F12 and approximately 5 mM OAC. In another specific embodiment, said medium comprises Stemspan® H3000 and approximately 5 mM OAC.
In certain embodiments, said second medium comprises cell growth medium comprising one or more of: human serum (eg, human serum AB), fetal bovine serum (FBS) or fetal calf serum (FCS), for example, 5% - 15% FCS v / v; IL-2, for example, 10 IU / mL at 1000 lU / mL; transferin, for example, 10 μ? /? t ?? at 50 μg mL; insulin, for example, 5 μg / mL at 20 μg mL; ethanolamine, for example, 5 x 10"4 to 5 x 10" 5 M; oleic acid, for example, 0.1 μg / mL at 5 μg / mL; linoleic acid, for example, 0.1 μg / mL at 5 μg / mL; palmitic acid, for example, 0.005 μg / ml_ at 2 μg / mL; bovine serum albumin (BSA), for example, 1 μ? / ???. at 5 μg / mL; and / or phytohemagglutinin, for example, 0.01 μg / mL to 1 μg / mL. In a specific embodiment, said second medium comprises IL-2. In a more specific embodiment, said second medium comprises cell growth medium comprising human serum, FBS or FCS, for example, 10% v / v, IL-2, transferin, insulin, ethanolamine, oleic acid, linoleic acid, palmitic acid. , bovine serum albumin (BSA) and / or phytohemagglutinin. In a more specific embodiment, said second medium comprises Iscove's modified Dulbecco's medium (IMDM), 10% human serum, FBS or FCS, 400 IU IL-2, 35 μg / mL insulin, 2 x 10"5 M ethanolamine, 1 μg / mL oleic acid, 1 μg / mL linoleic acid, 0.2 μ? / Gp? palmitic acid, 2.5 μg / mL BSA and 0.1 ng / mL phytohaemagglutinin In another specific modality, said culture in said second medium comprises cultivating using feeder cells, for example, K562 cells (e.g., K562 cells treated with mitomycin C) or PBMCs (e.g., PBMC treated with mitomycin C), e.g., at the time cells are initiated in said second medium, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 days later In certain embodiments, said medium comprises GBGM®, AIM-V®, X-VIVO ™ 10, X-VIVO ™ 15, OPTMIZER, STEMSPAN® H3000, CELLGRO COMPLETE ™, and / or DMEM: F12 In certain embodiments, said medium comprises one or more of O-acetyl-carnitine (also referred to as acetylcanitine, O-acetyl-L-carnit ina or OAC), or a compound that affects acetyl-CoA cyclization in mitodronia, thiazo- vivine, Y-27632, "py i ntegri n", Rho kinase inhibitors (ROCK), caspase inhibitors, or other anti-retroviral compounds / peptides. apoptotic, NOVA-RS (Sheffield Bio-Science) or other small molecule growth enhancers. In certain embodiments, said medium comprises nicotinamide. In certain embodiments, said medium comprises about 0.5 mM-10 mM OAC. In one embodiment, said medium comprises Stemspan® H3000, and / or DMEM: F12 and OAC, for example, about 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mM. In a specific embodiment, said medium comprises GBGM®. In another specific embodiment, said medium comprises DMEM: F12 and approximately 5 mM OAC. In another specific embodiment, said medium comprises Stemspan® H3000 and approximately 5 mM OAC.
In another specific embodiment, a method of producing a population of activated natural killer (NK) cells is provided, comprising: (a) planting a population of hematopoietic stem or progenitor cells in a first medium comprising interleukin-15 ( IL-15) and, optionally, one or more stem cell (SCF) and interleukin-7 (IL-7) factors, wherein said IL-15 and optionally SCF and IL-7 are not comprised within an undefined component of said medium, so that the population expands, and a plurality of hematopoietic stem or progenitor cells within said population of hematopoietic stem or progenitor cells differentiate into NK cells during said expansion; and (b) expanding the cells of step (a) in a second medium comprising interleukin-2 (IL-2), to produce a population of activated NK cells.
In another specific embodiment, a two-step method of producing a population of natural killer (NK) cells is provided herein, wherein a first step of said method comprises expanding a population of hematopoietic stem or progenitor cells into a first medium that comprises one or more of SCF, IL-7 and IL-15, and wherein said SCF, IL-7 and IL-15 are not comprised within an undefined component of said medium (e.g., serum), and wherein a plurality of hematopoietic stem or progenitor cells within said population of hematopoietic stem or progenitor cells differentiate into NK cells during said expansion; and wherein a second step of said method comprises expanding the cells from the first passage in a second medium comprising IL-2, to produce activated NK cells. In another specific embodiment, said first medium further comprises one or more of tyrosine kinase 3 ligand of the Fms type (FU3-L), thrombopoietin (Tpo), interleukin-2 (IL-2), and / or heparin. In another specific embodiment, said first medium further comprises about 5% -20% of fetal bovine serum or human serum. In another specific embodiment, the SCF is present at a concentration of about 1 to about 50 ng / mL in the first medium. In another specific embodiment, F 113-L is present at a concentration of about 1 to about 150 ng / mL in the first medium. In another specific embodiment, IL-2 is present at a concentration of about 50 to about 1500 IU / mL in the first medium. In another specific embodiment, IL-7 is present at a concentration of from about 1 to about 150 ng / mL in the first medium. In another specific embodiment, IL-15 is present at a concentration of from about 1 to about 150 ng / mL in the first medium. In another specific embodiment, the TPO is present at a concentration of about 1 to about 150 ng / mL in the first medium. In another specific embodiment, heparin is present at a concentration of about 0.1 to about 30 U / mL in the first medium. In another specific embodiment, the IL-2 in the second step is present at a concentration of about 50 to about 1500 IU / mL in the second medium. In another specific embodiment, said second means additionally comprises one or more of fetal calf serum (FCS), transferin, insulin, ethanolamine, oleic acid, linoleic acid, palmitic acid, bovine serum albumin (BSA) and phytohemagglutinin.
In certain specific embodiments, said hematopoietic stem or progenitor cells are cultured in said first medium during 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 or 28 days before said cultivation in said second medium. In certain other specific embodiments, said cells are cultured in said second medium during 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18 , 19, 20, 21, 22, 23, 24, 25, 26, 27 or 28 days. In a more specific embodiment, said hematopoietic stem or progenitor cells are cultured in said first medium for 21 days, and then cultured in said second medium for 21 days.
In addition, a population of natural killer cells produced by the two-step method described above, referred to herein as TSNK cells, is provided herein. In a specific embodiment, said NK cells (e.g., TSNK cells) are CD3"CD56 +." In a specific embodiment, said NK cells (e.g., TSNK cells) are CD3"CD56 + CD 16." In another specific embodiment, said NK cells (e.g., TSNK cells) are additionally CD94 + CD117 + In another specific embodiment, said NK cells (e.g., TSNK cells) are additionally CD161". In another specific embodiment, said NK cells (e.g., TSNK cells) are additionally NKG2D +. In another specific embodiment, said NK cells are additionally NKp464. In another specific embodiment, said NK cells are additionally CD226 +.
In certain modalities, more than 90%, 92%, 94%, 96% or 98% of said TSNK cells are CD56 + and CD16. "In some modalities, at least 80%, 82%, 84%, 86%, 88 % or 90% of said TSNK cells are CD3"and CD56 +. In other modalities, more than 90%, 92%, 94%, 96% or 98% of said TSNK cells are CD56 +, CD16 and CD3 \ In other modalities, at least 50%, 52%, 54%, 56% , 58% or 60% of said TSNK cells are NKG2D +. In other embodiments, less than 10%, 9%, 8%, 7%, 6%, 5%, 4% or 3% of said TSNK cells are NKB1 +. In certain other embodiments, less than 10%, 8%, 6%, 4% or 2% of said TSNK cells are CD56 + and CD16 +. In more specific modalities, at least 50%, 55%, 60%, 65% or 70% of said TSNK CD3"cells, CD56 + are NKp46 +, in other more specific modalities, at least 50%, 55%, 60 %, 65%, 70%, 75%, 80% or 85% of said TSNK CD3"cells, CD56 + are CD117 +. In other more specific embodiments, at least 20%, 25%, 30%, 35%, 40% or 45% of said TSNK CD3"cells, CD56 + are CD94 +, in other more specific modalities, at least 10%, 20 %, 25%, 30%, 35%, 40%, 45% or 50% of said TSNK CD3"cells, CD56 + are CD161." In other more specific modalities, at least 10%, 12%, 14%, 16 %, 18% or 20% of said TSNK CD3", CD56 * cells are CD226 +. In more specific embodiments, at least 20%, 25%, 30%, 35% or 40% of said TSNK CD3", CD56 + cells are CD7 +, in more specific embodiments, at least 30%, 35%, 40 %, 45%, 50%, 55% or 60% of said TSNK CD3", CD56 + are CD5 +.
In another aspect, the use of TSNK cells is provided herein to suppress tumor cell proliferation, treat viral infection or treat cancer, for example, blood cancers and solid tumors. In certain embodiments, the TSNK cells make contact with, or are used in combination with, an immunomodulatory compound, for example, an immunomodulatory compound described in section 6.2.1 below, or thalidomide.
In a specific embodiment, said cancer is a solid tumor. In another embodiment, said cancer is a blood cancer. In specific modalities, the cancer is glioblastoma, primary ductal carcinoma, leukemia, acute T cell leukemia, chronic myeloid lymphoma (CML), acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), lung carcinoma, colon adenocarcinoma, histiocytic lymphoma, colorectal carcinoma, colorectal adenocarcinoma, prostate cancer, multiple myeloma or retinoblastoma.
In another specific embodiment, hematopoietic cells, for example hematopoietic progenitor or stem cells, of which TSNK cells are produced, are obtained from perfusate of placenta, umbilical cord blood or peripheral blood. In another specific embodiment, hematopoietic cells, e.g., hematopoietic progenitor or stem cells, of which TSNK cells are produced, are combined placental perfusate and cord blood cells, e.g., cord blood from the same placenta as the perfused one In another specific embodiment, said umbilical cord blood is isolated from a placenta different from the placenta from which said placental perfusate is obtained. In certain embodiments, the combined cells can be obtained by pooling or combining cord blood and placental perfusate. In certain modalities, cord blood and placental perfusate are combined at a ratio of 100: 1, 95: 5, 90:10, 85:15, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45, 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85, 10:90, 5:95, 100: 1, 95: 1, 90: 1, 85: 1, 80: 1, 75: 1, 70: 1, 65: 1, 60: 1, 55: 1, 50: 1, 45: 1, 40: 1, 35: 1, 30: 1, 25: 1, 20: 1, 15: 1, 10: 1, 5: 1, 1: 1, 1: 5, 1:10, 1:15, 1:20, 1: 25, 1:30, 1:35, 1:40, 1:45, 1:50, 1:55, 1:60, 1:65, 1:70, 1:75, 1:80, 1:85, 1:90, 1:95, 1: 100, or similar by volume to obtain the combined cells. In a specific embodiment, cord blood and placental perfusate are combined at a ratio of 10: 1 to 1:10, 5: 1 to 1: 5, or 3: 1 to 1: 3. In other specific embodiments, cord blood and placental perfusate are combined at a ratio of 10: 1, 5: 1, 3: 1, 1: 1, 1: 3, 1: 5 or 1:10. In a more specific modality, cord blood and placental perfusate are combined at a ratio of 8.5: 1.5 (85%: 15%).
In certain modalities, cord blood and placental perfusate are combined at a ratio of 100: 1, 95: 5, 90:10, 85:15, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45, 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85, 10:90, 5:95, 100: 1, 95: 1, 90: 1, 85: 1, 80: 1, 75: 1, 70: 1, 65: 1, 60: 1, 55: 1, 50: 1, 45: 1, 40: 1, 35: 1, 30: 1, 25: 1, 20: 1, 15: 1, 10: 1, 5: 1, 1: 1, 1: 5, 1:10, 1:15, 1:20. 1:25, 1:30, 1:35, 1:40, 1:45, 1:50, 1:55, 1:60, 1:65, 1:70, 1:75, 1:80, 1: 85, 1:90, 1:95, 1: 100, or similar by total nucleated cell (TNC) content to obtain the combined cells. In a specific embodiment, cord blood and placental perfusate are combined at a ratio of 10: 1 to 10: 1, 5: 1 to 1: 5, or 3: 1 to 1: 3. In another specific embodiment, cord blood and placental perfusate are combined at a ratio of 10: 1, 5: 1, 3: 1, 1: 1, 1: 3, 1: 5 or 1:10.
In one embodiment, therefore, a method of treating an individual having cancer or a viral infection, which comprises administering to said individual an effective amount of isolated TSNK cells, is provided herein.
In a specific embodiment, the isolated TSNK cells have been treated with an immunomodulatory compound, for example, an immunomodulatory compound described in section 6.2.1 below, or thalidomide, prior to said administration. In another specific embodiment, the method comprises administering to the individual (1) an effective amount of isolated TSNK cells; and (2) an effective amount of an immunomodulatory compound or thalidomide. An "effective amount" in this context means an amount of TSNK cells, and optionally immunomodulatory compound or thalidomide, which results in a detectable improvement in one or more symptoms of said cancer or said infection, in comparison with an individual having said cancer or said infection that has not been administered said TSNK cells and, optionally, an immunomodulatory compound or thalidomide. In a specific embodiment, said immunomodulatory compound is lenalidomide or pomalidomide. In another embodiment, the method further comprises administering an anti-cancer compound to the individual, for example, one or more of the anti-cancer compounds described in section 6.8.3, below.
In another embodiment, a method of suppressing the proliferation of tumor cells comprising contacting the tumor cells with a therapeutically effective amount of TSNK cells is provided herein.
In a specific embodiment, the isolated TSNK cells have been treated with an immunomodulatory compound, for example, an immunomodulatory compound described in section 6.2.1 below, or thalidomide, prior to said contact. In another specific embodiment, the tumor cells are additionally contacted with an effective amount of an immunomodulatory compound, for example, an immunomodulatory compound described in section 6.2.1 below, or thalidomide. An "effective amount" in this context means an amount of TSNK cells, and optionally an immunomodulatory compound or thalidomide, which results in a detectable suppression of said tumor cells compared to an equivalent number of tumor cells not contacted with said TSNK cells, and optionally an immunomodulatory compound or Thalidomide In another specific embodiment, the method further comprises contacting the tumor cells with an effective amount of an anti-cancer compound, for example, an anti-cancer compound described in section 6.8.3 below.
In a specific embodiment of this method, the tumor cells are cancer lymphocytes. In another specific embodiment, the tumor cells are solid tumor cells. In another embodiment, the tumor cells are primary ductal carcinoma cells, leukemia cells, acute T cell leukemia cells, chronic myeloid lymphoma (CML) cells, acute myelogenous leukemia cells, chronic myelogenous leukemia (CML) cells, glioblastoma cells, lung carcinoma cells, colon adenocarcinoma cells, histiocytic lymphoma cells, multiple myeloma cells, retinoblastoma cells, colorectal carcinoma cells, prostate cancer cells, or colorectal adenocarcinoma cells. In another specific modality, said contact occurs in vitro. In another specific modality, said contact occurs in vivo. In a more specific modality, said in vivo contact occurs in a human.
In another aspect, herein is provided a method of treating an individual having multiple myeloma, comprising administering to the individual (1) lenalidomide; (2) melphalan; and (3) expanded NK cells, wherein said NK cells are effective to treat multiple myeloma in said individual. In a specific embodiment, said NK cells are NK cells of cord blood, or NK cells produced from cord blood hematopoietic cells, e.g., hematopoietic stem cells. In another embodiment, said NK cells have been produced by any of the methods described herein to produce NK cells, for example, to produce TSNK cells. In another embodiment, said NK cells have been expanded prior to said administration. In another embodiment, said lenalidomide, melphalan, and / or NK cells are administered separately from each other. In certain specific embodiments of the method of treating an individual with multiple myeloma, said NK cells are produced by a two-step method of producing a population of activated natural killer (NK) cells, wherein a first step of said method comprises expanding a population. of hematopoietic stem or progenitor cells in a first medium comprising one or more of the stem cell factor (SCF), interleukin-7 (I L-7) and interleukin-15 (IL-15), and wherein said SCF, IL-7 and IL-15 are not comprised within an undefined component of said medium, and wherein a plurality of hematopoietic stem or progenitor cells within said population of hematopoietic stem or progenitor cells differentiate into NK cells during said expansion; and wherein a second step of said method comprises expanding the cells from the first step into a second medium comprising interleukin-2 (IL-2), to produce activated NK cells.
In other specific modalities of the method of treating an individual with multiple myeloma, said NK cells are produced by a method comprising: (a) sowing a population of stem cells or hematopoietic progenitors in a first medium comprising interleukin-15 (IL-15) and, optionally, one or more of the stem cell factor (SCF) and interleukin-1 7 (IL-7), wherein said optional IL-15 and SCF and IL-7 are not comprised within an undefined component of said medium, such that the population expands, and a plurality of said population of stem cells or Hematopoietic progenitors differentiate into NK cells during said expansion; and (b) expanding the cells of step (a) in a second medium comprising interleukin-2 (IL-2), to produce a population of activated NK cells.
In another aspect, a method of treating an individual having chronic lymphocytic leukemia (CLL), which comprises administering to the individual a therapeutically effective dose of (1) lenalidomide, is provided; (2) melphalan; (3) Fludarabine; and (4) expanded NK cells, for example, TSNK cells, wherein said NK cells are effective to treat said CLL in said individual. In a specific embodiment, said NK cells are NK cells of cord blood, or NK cells produced from cord blood hematopoietic cells, e.g., hematopoietic stem cells. In another embodiment, said NK cells have been produced by any of the methods described herein to produce NK cells, for example, to produce TSNK cells. In a specific embodiment of any of the above methods, said NK cells have expanded for at least 10 days before said administration. In a specific embodiment of any of the above methods, said lenalidomide, melphalan, fludarabine and expanded NK cells are administered to said individual separately. In certain specific embodiments of the method of treating an individual with CLL, said NK cells are produced by a two-step method of producing a population of activated natural killer (NK) cells, wherein a first step of said method comprises expanding a population of hematopoietic stem or progenitor cells in a first medium comprising one or more of the stem cell factor (SCF), interleukin-7 (IL-7) and interleukin-15 (IL-15), and wherein said SCF, IL-7 and IL-15 are not comprised within an undefined component of said medium, and wherein a plurality of hematopoietic stem or progenitor cells within said population of hematopoietic stem or progenitor cells differentiate into NK cells during said expansion; and wherein a second step of said method comprises expanding the cells the first step into a second medium comprising interleukin-2 (IL-2), to produce activated NK cells.
In other specific embodiments of the method of treating an individual with CLL, said NK cells are produced by a method comprising: (a) planting a population of hematopoietic stem or progenitor cells in a first medium comprising interleukin-15 (IL-15) and, optionally, one or more of stem cell factor (SCF) and interleukin-7 (IL-7), wherein said optional IL-15 and SCF and IL-7 are not comprised within an undefined component of said medium , so that the population expands, and a plurality of hematopoietic stem or progenitor cells within said population of hematopoietic stem or progenitor cells differentiate into NK cells during said expansion; and (b) expanding the cells of step (a) in a second medium comprising interleukin-2 (IL-2), to produce a population of activated NK cells.
In one aspect, a method of cryopreservating a population of NK cells, e.g., TSNK cells, is provided herein. In one embodiment, said method comprises: (a) sowing a population of hematopoietic stem or progenitor cells in a first medium comprising interleukin-IL (IL-15) and, optionally, one or more of the stem cell factor (SCF) and interleukin-7 (IL-7), wherein said optional IL-15 and SCF and IL-7 are not comprised within an undefined component of said medium, so that the population expands, and a plurality of stem cells or Hematopoietic progenitors within said population of hematopoietic stem or progenitor cells differentiate into NK cells during said expansion; (b) expanding the cells of step (a) in a second medium comprising interleukin-2 (IL-2), to produce a population of activated NK cells, and (c) cryopreservating the NK cells of step (b) in a means of cryopreservation. In a specific embodiment, said step (c) further comprises (1) preparing a cell suspension solution; (2) adding cryopreservation medium to the cell suspension solution of step (1) to obtain cryopreserved cell suspension; (3) cooling the cryopreserved cell suspension of step (3) to obtain a cryopreserved sample; and (4) classify the cryopreserved sample below -80 ° C. In certain embodiments, the method includes any intermediate step between step (a) and (b), and between step (b) and (c).
In another embodiment, said method of cryopreservating a population of NK cells, for example, TSNK cells comprises: (a) expanding a population of hematopoietic stem or progenitor cells into a first medium comprising one or more of the stem cell factor (SCF) , IL-2, interleukin-7 (IL-7), interleukin-15 (IL-15) and heparin, and wherein said SCF, IL-2, IL-7 and IL-15 are not comprised within a non-component. defined from said medium, and wherein a plurality of hematopoietic stem or progenitor cells within said population of hematopoietic stem or progenitor cells differentiate into NK cells during said expansion; (b) expanding the cells of step (a) in a second medium comprising interleukin-2 (IL-2), to produce activated NK cells; and (c) cryopreservating the NK cells of step (b) in a cryopreservation medium. In a specific embodiment, said step (c) further comprises (1) preparing a cell suspension solution; (2) adding cryopreservation medium to the cell suspension solution of step (1) to obtain cryopreserved cell suspension; (3) cooling the cryopreserved cell suspension of step (3) to obtain a cryopreserved sample; and (4) store the cryopreserved sample below -80 ° C. In certain embodiments, the method includes any intermediate step between step (a) and (b), and between step (b) and (c), and / or no additional culture step before step (a).
In another specific embodiment, hematopoietic cells, e.g., hematopoietic progenitor or stem cells of which TSNK cells are produced express one or more of microRNAs hsa-mR-380, hsa-miR-512, hsa-miR-517 , hsa-miR-518c, hsa-miR-519b, and hsa-miR-520a at a detectably higher level than the natural killer cells of peripheral blood, as determined, for example, by quantitative real-time PCR (qRT- PCR).
In another specific embodiment of the above methods, said TSNK cells are contacted with an immunomodulatory compound or thalidomide in an amount and for a time sufficient for said natural killer cells to detectably express more granzyme B or perforin than an equivalent number of natural killer cells, for example, TSNK cells, not contacted with said immunomodulatory compound or thalidomide. In another specific embodiment, said TSNK cells are contacted with an immunomodulatory compound or thalidomide in an amount and for a time sufficient for said cells to detectably express more cytotoxicity to said tumor cells than an equivalent amount of natural killer cells, eg, TSNK cells. , not contacted with said immunomodulatory compound, for example, lenalidomide or pomalidomide, or with thalidomide. In other specific modality, said TSN cells express one or more of BAX, CCL5, CCR5, CSF2, FAS, GUSB, IL2RA or TNFRSF18 at a level higher than an equivalent number of natural killer cells, eg, TSNK cells, not contacted with said immunomodulatory compound or thalidomide. In another specific embodiment, said TSNK cells express one or more of ACTB, BAX, CCL2, CCL3, CCL5, CCR5, CSF1, CSF2, ECE1, FAS, GNLY, GUSB, GZMB, IL1A, IL2RA, IL8, IL10, LTA. , PRF1, PTGS2, SK1 and / or TBX21 at a level higher than an equivalent number of natural killer cells, e.g., TSNK cells, not contacted with said immunomodulatory compound or thalidomide.
In certain embodiments of the above tumor treatment or suppression methods, the TSNK cells are combined with other natural killer cells, eg, natural killer cells isolated from placental perfusate, umbilical cord blood or peripheral blood, or produced from hematopoietic cells by a different method In specific embodiments, the TSNK cells are combined with natural killer cells from another source, or made by a different method, in a ratio of approximately 100: 1, 95: 5, 90:10, 85:15, 80:20, 75 : 25, 70:30, 65:35, 60:40, 55:45, 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85 , 10:90, 5:95, 100: 1, 95: 1, 90: 1, 85: 1, 80: 1, 75: 1, 70: 1, 65: 1, 60: 1, 55: 1, 50 : 1, 45: 1, 40: 1, 35: 1, 30: 1, 25: 1, 20: 1, 15: 1, 10: 1, 5: 1, 1: 1, 1: 5, 1:10 1:15, 1:20, 1:25, 1:30, 1:35, 1:40, 1:45, 1:50, 1:55, 1:60, 1:65, 1:70, 1 : 75, 1:80, 1:85, 1:90, 1:95, 1: 100, or similar.
In another aspect, a comparison comprising isolating TSNK cells is provided herein. In a specific embodiment, said TSNKs are produced from hematopoietic cells, for example, hematopoietic progenitor or stem cells isolated from placental perfusate, umbilical cord blood, and / or peripheral blood. In another specific embodiment, said TSNK cells comprise at least 50% cells in the composition. In another specific embodiment, said TSNK cells comprise at least 80%, 85%, 90%, 95%, 98% or 99% of cells in the composition. In certain modalities, more than 90%, 92%, 94%, 96% or 98% of TSNK cells in said composition are CD56 + and CD16. "In other modalities, at least 80%, 82%, 84%, 86% , 88% or 90% of TSNK cells in said composition are CD3"and CD56 +. In other embodiments, at least 50%, 52%, 54%, 56%, 58% or 60% of said cells are NKG2D *. In other embodiments, less than 10%, 9%, 8%, 7%, 6%, 5%, 4% or 3% of said cells are NKB 1 +. In certain other embodiments, less than 10%, 8%, 6%, 4% or 2% of said TSNK cells are NKAT2 +. In certain other embodiments, less than 10%, 8%, 6%, 4% or 2% of said TSNK cells are CD56 + and CD16 +. In more specific modalities, at least 50%, 55%, 60%, 65% or 70% of said TSNK CD3"cells, CD56 + are NKp46 +, in other more specific modalities, at least 50%, 55%, 60 %, 65%, 70%, 75%, 80% or 85% of said TSNK CD3"cells, CD56 + are CD117 +. In other more specific modalities, at least 20%, 25%, 30%, 35%, 40% or 45% of said TSNK CD3"cells, CD56 + are CD94 +, in other more specific modalities, at least 10%, 12 %, 14%, 16%, 18% or 20% of said TSNK CD3, CD56 + cells are CD226 \ In more specific modalities, at least 20%, 25%, 30%, 35% or 40% of said TSNK CD3 cells ", CD56 + are CD7." In more specific modalities, at least 30%, 35%, 40%, 45%, 50%, 55% or 60% of said TSNK CD3, CD56 + cells are CD5 +.
In another specific embodiment, said TSNK CD56 +, CD16"cells are from a single individual, In a more specific embodiment, said natural killer cells CD56 +, CD16" comprise natural killer cells from at least two different individuals. In another specific embodiment, said TSNK cells have been contacted with an immunomodulatory compound or thalidomide in an amount and for a time sufficient for said TSNK cells to detectably express more granzyme B or perforin than an equivalent number of natural killer cells, i.e. TSNK, not contacted with said immunomodulatory compound or thalidomide. In another specific embodiment, said composition additionally comprises an immunomodulatory compound or thalidomide. In certain embodiments, the immunomodulatory compound is a compound described in section 6.2.1 below, for example, an amino-substituted isoindoline compound.
In another specific embodiment, the composition additionally comprises one or more anti-cancer compounds, for example, one or more of the anti-cancer compounds described in section 6.8.2 below.
In a more specific embodiment, the composition comprises TSNK cells and natural killer cells from another source, or made by another method. In a specific embodiment, said other source is placental blood and / or umbilical cord blood. In another specific embodiment, said other source is peripheral blood. In more specific modalities, TSNK cells are combined with natural killer cells from another source, or made by another method in a ratio of approximately 100: 1, 95: 5, 90:10, 85:15, 80:20, 75: 25, 70:30, 65:35, 60:40, 55:45, 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85, 10:90, 5:95, 100: 1, 95: 1, 90: 1, 85: 1, 80: 1, 75: 1, 70: 1, 65: 1, 60: 1, 55: 1, 50: 1, 45: 1, 40: 1, 35: 1, 30: 1, 25: 1, 20: 1, 15: 1, 10: 1, 5: 1, 1: 1, 1: 5, 1:10, 1:15, 1:20, 1:25, 1:30, 1:35, 1:40, 1:45, 1:50, 1:55, 1:60, 1:65, 1:70, 1: 75, 1:80, 1:85, 1:90, 1:95, 1: 100, or similar.
In another specific embodiment, the composition comprises TSNK cells and isolated placental perfusate or isolated placental perfusate cells. In a more specific embodiment, said placental perfusate is from the same individual as said TSNK cells. In another more specific embodiment, said placental perfusate comprises placental perfusate from an individual different from said TSNK cells. In another specific embodiment, all, or substantially all (eg, more than 90%, 95%, 98% or 99%) of the cells in said placental perfusate are fetal cells. In another specific embodiment, placental perfusate or placental perfusate cells comprise fetal and maternal cells. In a more specific embodiment, the fetal cells in said placental perfusate comprise less than about 90%, 80%, 70%, 60% or 50% of the cells in said perfusate. In another specific embodiment, said perfusate is obtained by passing a 0.9% NaCl solution through the placental vasculature. In another specific embodiment, said perfusate comprises a culture medium. In another specific embodiment, said perfusate has been treated to eliminate erythrocytes. In another specific embodiment, said composition comprises an immunomodulatory compound, for example, an immunomodulatory compound described in section 5.2.1.1 below, for example, an amino-substituted isoindoline compound. In another specific embodiment, the composition additionally comprises one or more anti-cancer compounds, for example, one or more of the anti-cancer compounds described in section 6.8.2 below.
In another specific embodiment, the composition comprises TSNK cells and placental perfusate cells. In a more specific embodiment, said placental perfusate cells are from the same individual as said TSNK cells. In another more specific embodiment, said placental perfusate cells are from a different individual than said TSNK cells. In another specific embodiment, the composition comprises isolated placental perfusate and isolated placental perfusate cells, wherein said placental perfusate and said isolated placental perfusate cells are of different individuals. In another modality more specific to any of the above embodiments comprising placental perfusate, said placental perfusate comprises placental perfusate of at least two individuals. In another more specific embodiment of any of the above embodiments comprising placental perfusate cells, said isolated placental perfusate cells are of at least two individuals. In another specific embodiment, said composition comprises an immunomodulatory compound. In another specific embodiment, the composition additionally comprises one or more anti-cancer compounds, for example, one or more of the anti-cancer compounds described in section 6.8.2, below. 4. 1 Definitions As used herein, the terms "immunomodulatory compound" and "I i D ™" do not encompass thalidomide.
As used herein, "lenalidomide" means 3- (4'-aminoisoindoline-1 -one) -1-piperidine-2,6-dione (name of Chemical Abstracts Service) or 2,6-piperidinedione, 3- (4-am ino- 1, 3-dihydro-1-oxo-2H-isoindol-2-yl) - (name of the International Union of Pure and Applied Chemistry (IUPAC)). As used herein, "pomalidomide" means 4-amino-2- (2,6-dioxopiperidin-3-yl) isoindol-1,3-dione.
As used herein, "multipotent", when referring to a cell, means that the cell has the ability to differentiate in a cell from another cell type. In certain embodiments, "a multipotent cell" is a cell that has the ability to grow in any subset of the mammalian body approximately 260 cell types. Unlike a pluripotent cell, a multipotent cell does not have the capacity to form all cell types.
As used herein, "feeder cells" refers to cells of a type that are co-cultured with cells of a second type, to provide an environment in which the cells of the second type can be maintained, and perhaps proliferate. Without being bound by any theory, the feeder cells may provide, for example, peptides, polypeptides, electrical signals, organic molecules (eg, steroids), nucleic acid molecules, growth factors (eg, bFGF), other factors (eg. example, cytokines), and metabolic nutrients to target cells. In certain embodiments, the feeder cells grow in a monolayer.
As used herein, "natural killer cell" or "NK cells" without further modification, includes natural killer cells from any tissue source.
As used herein, "TSNK" and "TSNK cells" refer to natural killer cells produced by the culture / expansion methods (eg, two-step method) described herein.
As used herein, "placental perfusate" means perfusion solution that has been passed through at least part of a placenta, for example, a human placenta, for example, through the placental vasculature, including a plurality of cells harvested by the perfusion solution during the passage through the placenta.
As used herein, "placental perfusate cells" means nucleated cells, e.g., total nucleated cells, isolated from, or that can be isolated from, placental perfusate.
As used herein, "tumor cell suppression", "suppression of tumor cell proliferation", and the like, includes reducing the growth of a population of tumor cells, for example, by killing one or more of the tumor cells in said population of tumor cells, for example, by contacting the population of tumor cells with, for example, TSNK cells or a population of cells comprising TSNK cells.
As used herein, the term "hematopoietic cells" includes hematopoietic stem cells and hematopoietic progenitor cells.
As used herein, the "undefined component" is a term of the art in the field of culture medium which refers to components whose constituents are generally not provided or quantified. Examples of an "undefined component" include, without limitation, human serum (e.g., AB human serum) and fetal serum (e.g., fetal bovine serum or fetal calf serum).
As used herein, "+", when used to indicate the presence of a particular cellular marker, means that the cellular marker is detectably present in fluorescent activated cell sorting over an isotype control; or it is background detectable in quantitative or semi-quantitative RT-PCR.
As used herein, when used to indicate the presence of a particular cellular marker, it means that the cellular marker is not detectably present in fluorescent activated cell sorting over an isotype control; or it is not detectable in the background in quantitative or semi-quantitative RT-PCR. 5. BRIEF DESCRIPTION OF THE FIGURES Figure 1: Fold expansion of differentiated NK cells from hematopoietic stem cells (HSCs) with various medium formulations. The error bars represent standard derivation of three donors. X axis: day (D) of culture. Y axis: fold expansion compared with day 0 (start of culture).
Figure 2: Phenotypic characterization of NK cells cultured with NK2A medium. Cells were labeled in triple with PE-antiCD56, FITC anti-CD3, PerCP-antiCD16. Horizontal, vertical lines: Fluorescent running level significantly background.
Figure 3: Fold expansion of NK cells cultured with NK2A (FF), NK2A (placental stem cells as feeder cells), NK2A (MSC as feeder cells) or two-stage NK medium. X axis: day (D) of culture. Y axis: fold expansion compared with day 0 (start of culture).
Figure 4: Cytotoxicity of NK cells cultured with NK2A (without feeder cells). Two-stage NK medium; NK2A with placental stem cells (PSC) adherent to tissue culture plastic CD34", CD10 \ CD105 \ CD200 + as feeder cells, NK2A with mesenchymal stem cells (MSC) as feeder cells, at post-culture initiation at day 45. The representative data of three donors are shown in Figure 4.
Figure 5: Phenotypic characterization of NK cells on day 41 (D41) of culture. Representative data of 3 individual donors are shown. X axis: percentage of NK cells, produced by the two-step method, which are CD3"CD56 +, CD16" CD56 + or CD16"CD56 +, or that express NKB1, NKG2D, NKp46, CD94, CD117, CD226, CD7 or CD5 Cells were cultured in NK2A (without feeder cells), two-stage NK medium, NK2A with placental stem cells (PSC) adherent to tissue culture plastic CD34", CD10 +, CD105 +, CD200 + as feeder cells, NK2A with mesenchymal stem cells (MSCs) derived from bone marrow as feeder cells, in the post-culture initiation on day 41.
Figure 6: Expression of CD94 and CD117 in the NK cell population CD56 + CD3- during NK culture in NK2A medium. The dominant population of CD56 + CD94 + CD 117 + cells was identified from NK cells cultured in NK2A medium, which can be distinguished from NK cells derived from embryonic stem cell (ESC) (CD56 + CD94 + CD117bai0"). of three donors are shown in Figure 6. X-axis: fluorescence of anti-CD94 labeled with phycoerythrin (PE) Y-axis: anti-CD117 fluorescence labeled with APC Horizontal, vertical lines: fluorescent label level significantly background D13 D20, D28, D35: initiation of cell culture post CD34 + days 13, 20, 28 and 35.
Figure 7: Effects of placental stem cells on cultured NK cells compared to MSC and NK2A medium alone. X axis: NK cells cultured in NK2A medium without a feeder layer (FF); NK cells cultured in NK2A medium with mesenchymal stem cells (MCS) derived from bone marrow as a feeder layer; or NK2A medium with placental stem cells (PDACs) adherent to tissue culture plastic CD10 +, CD34", CD105 +, CD200 + as a feeder layer Y-axis (left): cytotoxicity, expressed as a percentage of remaining tumor cells (1.0 = 100%), cytotoxicity indicated by open boxes, Y axis (right): expansion of NK cell fold using the two-step method, fold expansion expressed as asterisks.
Figures 8A-8B: Effects of relative relationships of umbilical cord blood (UCB) and human placental perfusate (HPP) on the purity of CD34 + post-thaw cells. Figure 8A: Effects of volumetric content of HPP (vol%) in purity of CD34 + Lin. "X axis: volumetric fraction of HPP in the UCB and grouped HPP (combination) Y axis: the percentage of CD34 + Lin cells". Figure 8B: Effects of HPP TNC content (TNC%) on CD34 + Lin purity "Y axis: the percentage of CD34 + Lin cells". 6. DETAILED DESCRIPTION Herein is provided a novel method of producing and expanding NK cells of hematopoietic cells, for example stem cells or hematopoietic progenitor cells. The hematopoietic cells used to produce the NK cells can be isolated from any source, for example, without limitation, placenta, umbilical cord blood, placental blood, peripheral blood, spleen or liver. In a certain embodiment, NK cells are produced from expanded hematopoietic cells, e.g., hematopoietic stem cells and / or hematopoietic progenitor cells. In one embodiment, hematopoietic cells are harvested from a source of said cells, for example, perfused from placenta, umbilical cord blood, peripheral blood, spleen, liver and / or bone marrow. In a specific embodiment, the hematopoietic cells are expanded and differentiated, continuously, in a first medium without the use of feeder cells. The cells are then cultured in a second medium in the presence of feeder cells. Said isolation, expansion and differentiation can be carried out in a central facility, which provides expanded hematopoietic cells for delivery for decentralized expansion and differentiation in points of use, for example, hospital, military base, military front, or similar. 6. 1. Hematopoietic cells The hematopoietic cells useful in the methods described herein can be any hematopoietic cell capable of differentiating into NK cells, for example, precursor cells, hematopoietic progenitor cells, hematopoietic stem cells, or the like. Hematopoietic cells can be obtained from tissue sources such as, for example, bone marrow, cord blood, placental blood, peripheral blood, liver or the like, or combinations thereof. Placental hematopoietic cells can be obtained. In a specific embodiment, the hematopoietic cells are obtained from placental perfusate. The hematopoietic cells of placental perfusate may comprise a mixture of fetal and maternal hematopoietic cells, for example, a mixture wherein maternal cells comprise more than 5% of the total number of hematopoietic cells. Preferably, the perfusate hematopoietic cells of placenta comprise at least about 90%, 95%, 98%, 99% or 99.5% fetal cells.
In another specific embodiment, hematopoietic cells, e.g. stem cells or hematopoietic progenitor cells, from which TSNK cells are produced, are obtained from perfusate of placenta, umbilical cord blood or peripheral blood. In another specific embodiment, hematopoietic cells, e.g., stem cells or hematopoietic progenitor cells, of which TSNK cells are produced, are combined cells from placental perfusate and cord blood, e.g., cord blood from the same placenta as the perfused one In another specific embodiment, said umbilical cord blood is isolated from a placenta different from the placenta from which said placental perfusate is obtained. In certain embodiments, the combined cells can be obtained by pooling or combining cord blood and placental perfusate. In certain modalities, cord blood and placental perfusate are combined at a ratio of 100: 1, 95: 5, 90:10, 85:15, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45, 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85, 10:90, 5:95, 100: 1, 95: 1, 90: 1, 85: 1, 80: 1, 75: 1, 70: 1, 65: 1, 60: 1, 55: 1, 50: 1, 45: 1, 40: 1, 35: 1, 30: 1, 25: 1, 20: 1, 15: 1, 10: 1, 5: 1, 1: 1, 1: 5, 1:10, 1:15, 1:20, 1: 25, 1:30, 1:35, 1:40, 1:45, 1:50, 1:55, 1:60, 1:65, 1:70, 1:75, 1:80, 1:85, 1:90, 1:95, 1: 100, or similar by volume to obtain the combined cells. In a specific embodiment, cord blood and placental perfusate are combined at a ratio of 10: 1 to 1:10, 5: 1 to 1: 5, or 3: 1 to 1: 3.
In another specific embodiment, cord blood and placental perfusate are combined at a ratio of 10: 1, 5: 1, 3: 1, 1: 1, 1: 3, 1: 5 or 1:10. In a more specific modality, cord blood and placental perfusate are combined at a ratio of 8.5: 1.5 (85%: 15%).
In certain modalities, cord blood and placental perfusate are combined at a ratio of 100: 1, 95: 5, 90:10, 85:15, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45, 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85, 10:90, 5:95, 100: 1, 95: 1, 90: 1, 85: 1, 80: 1, 75: 1, 70: 1, 65: 1, 60: 1, 55: 1, 50: 1, 45: 1, 40: 1, 35: 1, 30: 1, 25: 1, 20: 1, 15: 1, 10: 1, 5: 1, 1: 1, 1: 5, 1:10, 1:15, 1:20, 1: 25, 1:30, 1:35, 1:40, 1:45, 1:50, 1:55, 1:60, 1:65, 1:70, 1:75, 1:80, 1:85, 1:90, 1:95, 1: 100, or similar by total nucleated cell (TNC) content to obtain the combined cells. In a specific embodiment, cord blood and placental perfusate are combined at a ratio of 10: 1 to 10: 1, 5: 1 to 1: 5, or 3: 1 to 1: 3. In another specific embodiment, cord blood and placental perfusate are combined at a ratio of 10: 1, 5: 1, 3: 1, 1: 1, 1: 3, 1: 5 or 1:10.
In another specific embodiment, the hematopoietic cells, for example, stem cells or hematopoietic progenitor cells from which said TSNK cells are produced are from umbilical cord blood and perfused from placenta, but wherein said umbilical cord blood is isolated from a placenta different from the placenta from which said placental perfusate is obtained.
In certain embodiments, the hematopoietic cells are CD34 + cells. In specific embodiments, the hematopoietic cells useful in the methods described herein are CD34 + CD38 + or CD34 + CD38. "In a more specific embodiment, the hematopoietic cells are CD34 + CD38 ~ Lin ~ In another specific embodiment, the hematopoietic cells are one or more of CD2", CD3, CD11b ~, CD11c", CD14", CD16", CD19", CD24", CD56-, CD66b "and / or glycophorin A." In another specific embodiment, the hematopoietic cells are CD2 ', CD3", CD11b", CD11c ", CD14", CD16", CD19", CD24", CD56", CD66b "and glycophorin A." In another more specific modality, the hematopoietic cells are CD34 + CD38"CD33" CD117. " In another more specific embodiment, the hematopoietic cells are CD34 + CD38"CD33" CD 117 CD25 CD36".
In another embodiment, the hematopoietic cells are CD45 +. In another specific embodiment, the hematopoietic cells are CD34 + CD45 +. In another embodiment, the hematopoietic cell is Thy-1 +. In a specific embodiment, the hematopoietic cell is CD34 + Thy-1 +. In another embodiment, the hematopoietic cells are CD133 +. In specific embodiments, the hematopoietic cells are CD34 + CD133 + or CD133 + Thy-1 \ In another specific embodiment, the CD34 + hematopoietic cells are CXCR4 +. In another specific modality, the CD34 + hematopoietic cells are CXCR4. "In another embodiment, the hematopoietic cells are positive for KDR (vascular growth factor receptor 2.) In specific modalities, the hematopoietic cells are CD34 + KDR +, CD133 + KDR or Thy-1 + KDR + In certain other modalities, the hematopoietic cells are positive for aldehyde dehydrogenase (ALDH +), for example, the cells are CD34 + ALDH \ In certain other embodiments, CD34 + cells are CD45. "In specific embodiments, CD34 + cells, eg, CD34 +, CD45 cells" express one or more, or all, of miRNAs hsa-miR-380, hsa-miR -512, hsa-miR-517, hsa-miR-518c, hsa-mR-519b, and / or hsa-miR-520a.
In certain embodiments, the hematopoietic cells are CD34. Hematopoietic cells may also lack certain markers that indicate lineage involvement, or a lack of developmental ingenuity. For example, in another modality, the hematopoietic cells are HLA-DR. "In specific modalities, the hematopoietic cells are CD34 + HLA-DR", CD133 + HLA-DR ", Thy-1 + HLA-DR" or ALDH + HLA -DR. "In another embodiment, the hematopoietic cells are negative for one or more, preferably all, of the lineage markers CD2, CD3, CD11c, CD14c, CD14, CD16, CD19, CD24, CD56b, and glycophorin A.
In this manner, hematopoietic cells can be selected for use in the methods described herein on the basis of the presence of markers indicating an undifferentiated state, or on the basis of the absence of lineage markers indicating that at least one occurrence has occurred. some lineage differentiation. Methods of isolating cells, including hematopoietic cells, on the basis of the presence or absence of specific markers are discussed in detail, for example, in section 6.1.2, below.
The hematopoietic cells used in the methods provided herein may be a substantially homogeneous population, for example, a population comprising at least about 95%, at least about 98% or at least about 99% of a single hematopoietic cell. tissue source, or a population comprising hematopoietic cells that exhibit the same cellular markers associated with hematopoietic cells. For example, in various embodiments, the hematopoietic cells may comprise at least about 95%, 98% or 99% of hematopoietic cells of bone marrow, cord blood, placental blood, peripheral blood, or placenta, for example, perfused with placenta.
The hematopoietic cells used in the methods provided herein can be obtained from a single individual, for example, from a single placenta, or from a plurality of individuals, for example, they can be grouped. Where the hematopoietic cells are obtained from a plurality of individuals and grouped, the hematopoietic cells can be obtained from the same source of tissue. Thus, in various embodiments, the grouped hematopoietic cells are all placental, for example, placental perfused, all peripheral blood, all umbilical cord blood, all peripheral blood, and the like.
The hematopoietic cells used in the methods described herein, in certain embodiments, may comprise hematopoietic cells from two or more tissue sources. For example, in certain embodiments, when hematopoietic cells from two or more sources are combined for use in the methods herein, a plurality of the hematopoietic cells used to produce TSNK cells comprise placental hematopoietic cells, for example, placental perfusate. In several embodiments, the hematopoietic cells used to produce TSNK cells comprise hematopoietic cells from placenta and cord blood; of placenta and peripheral blood; of placenta and peripheral blood; or placenta and bone marrow. In a preferred embodiment, the hematopoietic cells comprise placental perfusate hematopoietic cells in combination with cord blood hematopoietic cells, wherein cord blood and placenta are from the same individual, i.e., where perfusate and blood are matched. cord. In embodiments wherein the hematopoietic cells comprise hematopoietic cells from two tissue sources, the hematopoietic cells of the sources can be combined in a ratio of, for example, 1:10, 2: 9, 3: 8, 4: 7, 5 : 6, 6: 5, 7: 4, 8: 3, 9: 2, 1:10, 1: 9, 1: 8, 1: 7, 1: 6, 1: 5, 1: 4, 1: 3 , 1: 2, 1: 1, 2: 1, 3: 1, 4: 1, 5: 1, 6: 1, 7: 1, 8: 1 or 9: 1. 6. 1.1 Hematopoietic Placental Stem Cells In certain embodiments, the hematopoietic cells used in the methods provided herein are placental hematopoietic cells. As used herein, "placental hematopoietic cells" means hematopoietic cells obtained from the placenta itself, and not from placental blood or umbilical cord blood. In one embodiment, the placental hematopoietic cells are CD34 +. In a specific embodiment, the placental hematopoietic cells are predominantly (eg, at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 98%) CD34 + CD38 cells. "In another specific embodiment, placental hematopoietic cells are predominantly (eg, at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%). %, 90%, 95% or 98%) CD34 + CD38 + cells Placental hematopoietic cells can be obtained from a postpartum mammalian placenta (eg, human) by any means known to those skilled in the art, by example, by perfusion.
In another embodiment, the placental hematopoietic cell is CD45. "In a specific modality, the hematopoietic cell is CD34 + CD45." In another specific embodiment, the placental hematopoietic cells are CD34 + CD45 +. 6. 2. Production of Natural Killer Cells The production of NK cells by the present method comprises expanding a population of hematopoietic cells. During cell expansion, a plurality of hematopoietic cells within the hematopoietic cell population differentiate into NK cells.
In one embodiment, a method of producing a population of activated natural killer (NK) cells is provided, comprising: (a) seeding a population of hematopoietic stem or progenitor cells in a first medium comprising interleukin-15 (IL) - 15) and, optionally, one or more of the stem cell factor (SCF) and interleukin-7 (IL-7), wherein said optional IL-15 and SCF and IL-7 are not included within an undefined component of said medium, so that the population expands, and a plurality of hematopoietic stem or progenitor cells within said population of hematopoietic stem or progenitor cells differentiate into NK cells during said expansion; and (b) expanding the cells of step (a) in a second medium comprising interleukin-2 (IL-2), to produce a population of activated NK cells.
In another embodiment, the NK cells provided herein are produced by a two-step process of NK cell expansion / differentiation and maturation. The first and second steps involve growing the cells in medium with a unique combination of cellular factors. In certain embodiments, the process involves (a) culturing and expanding a population of hematopoietic cells in a first medium, wherein a plurality of hematopoietic stem or progenitor cells within the population of hematopoietic cells differentiate into NK cells; and (b) expanding the NK cells of step (a) in a second medium, wherein the NK cells expand further and differentiate, and where the NK cells are matured (eg, activated or otherwise possessing cytotoxic activity) . In certain embodiments, the method includes any intermediate step between step (a) and step (b), no additional culture step before step (a), and / or no additional step (eg, maturation step) after the step (b). 6. 2.1. First Growing Step In certain embodiments, the methods provided herein comprise a first step of culturing and expanding a population of hematopoietic cells in a first medium, wherein a plurality of hematopoietic stem or progenitor cells within the population of hematopoietic cells differentiate into NK cells. .
Without being limited by any parameter, mechanism or theory, culture of the hematopoietic cells as provided herein results in continuous expansion of the hematopoietic cells and differentiation of NK cells from said cells. In certain embodiments, the hematopoietic cells, e.g., stem cells or progenitor cells, used in the methods provided herein expand and differentiate in the first step using a feeder layer. In other embodiments, the hematopoietic cells, e.g., stem cells or progenitor cells, expand and differentiate in the first step without the use of a feeder layer.
The independent expansion and differentiation of hematopoietic cell feeder cells can occur in any container compatible with culture and cell expansion, for example, bottle, tube, beaker, dish, multi-well plate, bag or the like. In a specific embodiment, the independent expansion of the hematopoietic cell feeder cell occurs in a bag, for example, a flexible, gas-permeable fluorocarbon culture bag (for example, from American Fluoroseal). In a specific embodiment, the container in which the hematopoietic cells expand is suitable for delivery, for example, to a site such as a hospital or military zone where the expanded NK cells expand and further differentiate.
In certain embodiments, the hematopoietic cells expand and differentiate, for example, in a continuous mode, in a first culture medium. In one embodiment, the first culture medium is a free medium of animal component. Exemplary animal component free media useful in the methods provided herein include, but are not limited to, Eagle Basal Medium (BME), Dulbecco's Modified Eagle Medium (DMEM), Glasgow Minimum Essential Medium (GMEM), Medium modified Dulbecco's Eagle / F-12 Ham nutrient mixture (DMEM / F-12), minimum essential medium (E), Iscove's modified Dulbecco's medium (IMDM), Ham's F-10 nutrient mixture (F-10 Ham), mixture of F-12 Ham nutrient (Ham F-12), RPMI-1640 medium, Wílliam medium E, STEMSPAN® (Cat. Stem Cell Technologies, Vancouver, Canada), basal growth medium Glycostem Basal Growth Medium (GBGM®), AIM-V® media (Invitrogen), X-VIVO ™ 10 (Lonza), X-VIVO ™ 15 (Lonza), OPTIMIZER (Invitrogen), STEMSPAN® H3000 (STEMCELL Technologies), CELLGRO COMPLETE ™ (Mediatech), or any modified variant or combinations thereof.
In preferred embodiments, the first culture medium comprises one or more medium supplements (e.g., nutrients, cytokines and / or factors). Suitable medium supplements for use in the methods provided herein include, for example, without limitation, serum such as human serum AB, fetal bovine serum (FBS) or fetal calf serum (FCS), vitamins, bovine serum albumin ( BSA), amino acids (e.g., L-glutamine), fatty acids (e.g., oleic acid, linoleic acid or palmitic acid), insulin (e.g., recombinant human insulin), transferin (human saturated iron transferrin), β- mercaptoethanol, stem cell factor (SCF), FMS tyrosine kinase 3 ligand (FU3-L), cytokines such as interleukin-2 (IL-2), interleukin-7 (IL-7), interleukin -15 (IL-15), thrombopoietin (Tpo), heparin, or O-acetyl-carnitine (also referred to as acetylcarnitine, O-acetyl-L-carnitine or OAC). In a specific embodiment, the medium used comprises human serum AB. In another specific embodiment, the medium used herein comprises FBS. In another specific embodiment, the medium used herein comprises OAC.
In certain embodiments, the first medium does not comprise one or more of, granulocyte colony stimulating factor (G-CSF), granulocyte / macrophage colony stimulating factor (GM-CSF), interleukin-6 (IL-6), protein 1 a macrophage inflammatory (MIP1a), or leukemia inhibitory factor (LIF).
Thus, in one aspect, a two-step method of producing NK cells is provided herein, wherein said first step comprises expanding and differentiating a population of hematopoietic cells in a first culture medium in the absence of feeder cells, in wherein a plurality of hematopoietic cells within said population of hematopoietic cells differentiate into NK cells during said expansion, and wherein the medium comprises SCF at a concentration of about 1 to about 150 ng / mL, IL-2 at a concentration of about 50 to about 1500 IU / mL, IL-7 at a concentration of about 1 to about 150 ng / mL, IL-15 at a concentration of about 1 to about 150 ng / mL and heparin at a concentration of about from 0.1 to about 30 lU / mL, and where said SCF, IL-2, IL-7, IL-15 and heparin are not comprised within an undefined component of said medium (e.g., serum). In certain embodiments, said medium comprises one or more of O-acetyl-carnitine (also referred to as acetylcarnitine, O-acetyl-L-carnitine or OAC), or a compound that affects the acetyl-CoA cycle in mitodronia, thiazo-vivine, and -27632, piintegrin, Rho kinase inhibitors (ROCK), caspase inhibitors or other anti-apoptotic compounds / peptides, NOVA-RS (Sheffield Bio-Science) or other small molecule growth enhancers. In certain modalitiessaid medium comprises nicotinamide. In certain embodiments, said medium comprises approximately 0.5 mM-10 mM OAC. In one embodiment, said medium comprises Stemspan® H3000, and / or DM MS: F 12 and approximately 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mM OAC. In a specific embodiment of the method, said medium is GBGM®. In another specific embodiment, said medium comprises Stemspan® H3000 and approximately 5 mM OAC. In another specific embodiment, said medium comprises DMEM: F12 and approximately 5 nM OAC. The OAC may be added at any time during the culture methods provided herein. In certain modalities, said OAC is added to the first means on day 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 of the culture. In a specific embodiment, said OAC is added to the first medium on day 7 of the first culture step. In a more specific embodiment, said OAC is added to the first medium on day 7 of the culture and is present in the first and second cultivation steps. In certain embodiments, said OAC is added to the second medium and / or during the second culture step. In some embodiments, said OAC is added to the second medium on day 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 of the culture.
In another specific embodiment, said medium is IMDM supplemented with approximately 5-20% BSA, approximately 1-10 μg / mL recombinant human insulin, approximately 10-50 μg / mL human iron transferrin saturated and approximately 10-50 μ? β-mercaptoethanol. In another specific embodiment, said medium does not comprise one or more, or none, of IL-11, IL-3, homeobox-B4 (HoxB4), and / or methylcellulose.
In other specific embodiments, said medium comprises SCF at a concentration of from about 0.1 to about 500 ng / mL; from about 5 to about 100 ng / mL; or approximately 20 ng / mL. In other specific embodiments, said medium comprises IL-2 at a concentration of from about 10 to about 2000 lU / mL; or from about 100 to about 500 IU / mL; or approximately 200 lU / mL. In other specific embodiments, said medium comprises IL-7 at a concentration of from about 0.1 to about 500 ng / mL; from about 5 to about 100 ng / mL; or approximately 20 ng / mL. In other specific embodiments, said medium comprises IL-15 at a concentration of from about 0.1 to about 500 ng / mL; from about 5 to about 100 ng / mL; or approximately 10 ng / mL. In other specific embodiments, said medium comprises heparin at a concentration of about 0.05 to about 100 IU / mL; or from about 0.5 to about 20 U / ml; or approximately 1.5 U / mL.
Still in another specific embodiment of the method, said medium further comprises Fms-type tyrosine kinase 3 ligand (Flt-3L) at a concentration of from about 1 to about 150 ng / mL, thrombopoietin (Tpo) at a concentration of about 1. at about 150 ng / mL, or a combination of about 0.1 to about 500 ng / mL; from about 5 to about 100 ng / mL; or approximately 20 ng / mL. In other specific embodiments, said medium comprises TPO at a concentration of from about 0.1 to about 500 ng / mL; from about 5 to about 100 ng / mL; or approximately 20 ng / mL.
In a more specific mode of the method, the first culture medium is GBGM®, which comprises approximately 20 ng / mL SCF, approximately 20 ng / mL IL-7, approximately 10 ng / mL IL-15. In another more specific embodiment of the method, the first culture medium is GBGM®, which comprises approximately 20 ng / mL SCF, approximately 20 ng / mL FH3-L, approximately 200 lU / mL IL-2, approximately 20 ng / mL IL -7, approximately 10 ng / mL IL-15, approximately 20 ng / mL Tpo, and approximately 1.5 U / mL heparin. In another specific embodiment, said first culture medium further comprises 10% human serum (e.g., human serum AB) or fetal serum (e.g., FBS).
In another embodiment, the hematopoietic cells are expanded by culturing said cells, for example, in said first medium, in contact with an immunomodulatory compound, for example, a TNF-α inhibitor compound, for a time and in an amount sufficient to cause a detectable increase in the proliferation of hematopoietic cells over a given time, compared to an equivalent number of hematopoietic cells not contacted with the immunomodulatory compound. See, for example, patent application publication of E.U.A. No. 2003/0235909, the description of which is incorporated herein by reference in its entirety. In certain embodiments, the immunomodulatory compound is an amino-substituted isoindoline. In a preferred embodiment, the immunomodulatory compound is 3- (4-amino-1-oxo-1,3-dihydro-isoindol-2-yl) -piperidine-2,6-dione; 3- (4'-aminoisoindolin-1'-one) -1-piperidine-2,6-dione; 4- (amino) -2- (2,6-dioxy (3-piperidyl)) - isoindoline-1,3-dione; or 4-amino-2- (2,6-dioxopiperidin-3-yl) isoindol-1,3-dione. In another preferred embodiment, the immunomodulatory compound is pomalidomide or lenalidomide. In another embodiment, said immunomodulatory compound is a compound having the structure wherein one of X and Y is C = 0, the other of X and Y is C = 0 or CH2, and R2 is hydrogen or lower alkyl, or a pharmaceutically acceptable salt, hydrate, solvate, clathrate, enantiomer, diastereomer, racemate or mixture of stereoisomers thereof. In another embodiment, said immunomodulatory compound is a compound having the structure where one of X and Y is C = 0 and the other is CH2 or C = 0; R is H, (d-C8) alkyl, (C3-C7) cycloalkyl, (C2-C8) alkenyl, (C2-C8) alkynyl, benzyl, aryl, (C0-C4) alkyl -heterocycloalkyl (d-Ce), (C0-C4) alkyl -heteroaryl (C2-C5), C (0) R3, C (S) R3, C (0) OR4, alkyl (d -C8) -N (R6) 2, alkyl (d-C8) ) -OR5, (Ci-C8) alkyl -C (0) OR5, C (0) NHR3, C (S) NHR3, C (0) NR3R3 ', C (S) NR3R3' or alkyl (d-C8) - 0 (CO) R5; R2 is H, F, benzyl, alkyl (d-C8), alkenyl (C2-C8), or alkynyl (C2-C8); R3 and R3 are independently alkyl (d-C8), cycloalkyl (C3-C7), alkenyl (C2-C8), alkynyl (C2-C8), benzyl, aryl, alkyl (C0-d) -heterocycloalkyl (C ^ -d) ), alkyl (C0-C) -heteroaryl (C2-C5), (C0-C8) alkyl -N (R6) 2, alkyl (d-C8) -OR5, alkyl (d-C8) -C (0) OR5 , alkyl (d-C8) -0 (CO) R5 or C (0) OR5; R4 is alkyl (C ^ -C ^), alkenyl (C2-C8), alkynyl (C2-C8), alkyl (C-C4) -OR5, benzyl, aryl, (C0-C4) alkyl -heterocycloalkyl (Ci-Ce) ) or (C0-C4) alkyl-heteroaryl (C2-C5); R5 is (Ci-C8) alkyl, (C2-C8) alkenyl, (C2-C8) alkynyl, benzyl, aryl or (C2-C5) heteroaryl; each occurrence of R6 is independently H, (Ci-C8) alkyl, (C2-C8) alkenyl, (C2-C8) alkynyl, benzyl, aryl, (C2-C5) heteroaryl or (C0-C8) alkyl-C (O ) O-R5 or the R6 groups can be combined to form a heterocycloalkyl group; n is 0 or 1; Y * represents a chiral carbon center; or a salt, hydrate, solvate, clathrate, enantiomer, diastereomer, racemate, or mixture of stereoisomers thereof. In another embodiment, said immunomodulator compound is a compound having the structure where: one of X and Y is C = 0 and the other is CH2 or C = 0; R is H or CH2OCOR '; (i) each of R1, R2, R3 or R4, independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms or (ii) one of R1, R2, R3 or R4 is nitro or -NHR5 and the remainder of R \ R2, R3 or R4 are hydrogen; R5 is hydrogen or alkyl of 1 to 8 carbon atoms R6 is hydrogen, alkyl of 1 to 8 carbon atoms, benzo, chloro or fluoro; R 'is R7-CHR10-N (R8R9); R7 is m-phenylene or p-phenylene or - (CnH2n) - wherein n has a value of 0 to 4; each of R8 and R9 taken independently of the other is hydrogen or alkyl of 1 to 8 carbon atoms, or R8 and R9 taken together are tetramethylene, pentamethylene, hexamethylene, or -CH2CH2X1CH2CH2- wherein X1 is -O-, -S- or -NH-; R10 is hydrogen, alkyl of 8 carbon atoms, or phenyl; Y * represents a chiral carbon center; or a salt, hydrate, solvate, clathrate, enantiomer, diastereomer, racemate, or mixture of stereoisomers thereof.
In a specific embodiment, the expansion of the hematopoietic cells is performed in IMDM supplemented with 20% BITS (bovine serum albumin, recombinant human insulin and transferin), SCF, ligand Flt-3, IL-3 and 4- (amino) - 2- (2,5-dioxo (3-piperidyl)) - isoindoline-1,3-dione (10 μ ?? in 0.05% DMSO). In a more specific embodiment, approximately 5 x 10 7 hematopoietic cells, eg, CD34 + cells, expand in the medium from about 5 x 10 10 cells to about 5 x 10 12 cells, which are resuspended in 100 ml_ of IMDM to produce a population of expanded hematopoietic cells. The population of expanded hematopoietic cells is preferably cryopreserved to facilitate shipping.
In several specific modalities, at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% of the hematopoietic cells they differentiate to NK cells.
In certain embodiments, the method of expansion and differentiation of hematopoietic cells, as described herein, comprises maintaining the cell population comprising said hematopoietic cells at between about 2 x 10 4 and about 2 x 10 5 cells per milliliter during the expansion and differentiation. In certain other embodiments, the method of expansion and differentiation of hematopoietic cells, as described herein, comprises maintaining the cell population comprising said hematopoietic cells at no more than about 1 x 10 5 cells per milliliter.
The time for expansion and differentiation of hematopoietic cells into NK cells can be, for example, from about 3 days to about 120 days. In one embodiment, the differentiation time is around 7 days to about 75 days. In another modality, the differentiation time is around 14 days to approximately 50 days. In a specific modality, the differentiation time is around 21 days to about 28 days. 6. 2.2. Second step In the method provided herein, the hematopoietic cells, e.g., stem cells or progenitor cells, and natural killer cells, which result from the first step, expand further and differentiate in a second step, e.g., without the use of a layer. feeder or in the presence of feeder cells. The culture of the cells as provided herein results in continuous expansion, differentiation as well as maturation of the NK cells of the first step. In the second step, the NK cells are expanded, differentiated and matured, in a continuous mode, in a second culture medium, for example, comprising different cytokines and / or bioactive molecules than said first medium. In certain embodiments, the second culture medium is a free medium of animal component. Exemplary animal component free cell culture media are described in section 6.2.1, above.
Thus, in one aspect, a method of producing NK cells is provided herein, comprising expanding the NK cells of the first step, described above, into a second medium in the presence of feeder cells and in contact with interleukin-2 (IL -2). In specific embodiments, said second medium comprises cell growth medium comprising IL-2, for example, 10 IU / mL at 1000 IU / mL, and one or more of: human serum (e.g., human AB serum), bovine serum fetal (FBS) or fetal calf serum (FCS), for example, 5% - 15% FCS v / v; transferin, for example, 10 μ? / mL at 50 μ? / mL; insulin, for example, 5 μg |. { ? L · a 20 μ? /? T ?? -; ethanolamine, for example, 5 x 10"to 5 x 10" 5 M; oleic acid, for example, 0.1 μ? /? t ?? at 5 μ? /? t ?? -; linoleic acid, for example, 0.1 μg / μl to 5 μl / mL ·: palmitic acid, for example, 0.05 μg / G. at 2 μ? / ??? -; bovine serum albumin (BSA), for example, 1 μ9 /? to 5 μ9 / ??; and / or phytohaemagglutinin, for example, 0.01 μ9 / λ "_ to 1 μ9 /?" - In a more specific embodiment, said second medium comprises cell growth medium comprising FBS or FCS, eg, 10% FCS v / v, IL-2, transferin, insulin, ethanolamine, oleic acid, linoleic acid, palmitic acid, bovine serum albumin (BSA) and phytohemagglutinin In a more specific embodiment, said second medium comprises modified Dulbecco's medium from Iscove (IMDM), 10% FBS or FCS, 400 IU IL-2, 35 μg / mL transferin, 5 μg / mL insulin, 2 x 10 ~ 5 M ethanolamine, 1 μ? / ??? - oleic acid, 1 μ? / mL linoleic acid (Sigma-Aldrich), 0.2 μgglL · palmitic acid (Sigma-Aldrich), 2.5 μg / mL BSA (Sigma-Aldrich) and 0.1 μ9 / (? _ phytohemagglutinin.
In certain embodiments, the second medium does not comprise one or more of, granulocyte colony stimulating factor (G-CSF), granulocyte / macrophage colony stimulating factor (GM-CSF), interleukin-6 (IL-6), protein 1 a macrophage inflammatory (MIP1 a), or leukemia inhibitory factor (LIF).
In addition to the method, any of the means described above as compositions are provided herein.
Feeding cells, when used, of various cell types can be established. Examples of these cell types include, without limitation, fibroblasts, stem cells (e.g., placental stem cells adhering to tissue culture), fibroblasts (e.g., peripheral blood mononuclear cells (PBMC)), and cancer cells (e.g. , chronic myelogenous leukemia (CML) cells such as K562). In a specific embodiment, said culture in said second means comprises cultivating using feeder cells, for example, K562 cells and / or peripheral blood mononuclear cells (PBMCs), for example, at the time when cells initiate in said second medium, , 2, 3, 4, 5, 6, 7, 8, 9 or 10 days later. In certain embodiments, the feeder cells optionally are of a different species as the cells they support. For example, human NK cells can be supported by mouse embryonic fibroblasts (primary culture or a telomerized line).
In certain embodiments, the feeder cells are optionally inactivated by irradiation (e.g., irradiation?) Or treatment with an anti-mitotic agent such as mitomycin C, to prevent them from overtaking the cells they are supporting, but allow the synthesis of important factors that support NK cells. For example, cells can be irradiated at a dose to inhibit proliferation but allow synthesis of important factors that They support human embryonic stem cells (hES) (approximately 4000 rads gamma irradiation).
The culture of NK cells for the second step can occur in any container compatible with culture and cell expansion, for example, bottle, tube, beaker, dish, multi-well plate, bag or the like. In a specific embodiment, the NK cell feeder-dependent culture occurs in a bag, eg, gas-permeable fluorocarbon flexible culture bag (e.g., from American Fluoroseal). In a specific embodiment, the container in which the NK cells are cultured is suitable for shipping, for example, to a site such as a hospital or military zone where the expanded NK cells expand more, differentiate and mature.
The differentiation of cells from step 1 into TSNK cells can be evaluated by detecting specific NK cell markers, for example, by flow cytometry. Specific NK cell markers include, but are not limited to, CD56, CD94, CD117 and NKp46. Differentiation can also be assessed by the morphological characteristics of NK cells, for example, large size, high protein synthesis activity in the abundant endoplasmic reticulum (ER) and / or preformed granules.
The time for expansion and differentiation of cells from step 1 into TSNK cells can be, for example, from about 3 days to about 120 days. In one embodiment, the differentiation time is around 7 days to about 75 days.
In another modality, the differentiation time is around 14 days to approximately 50 days. In a specific modality, the differentiation time is around 10 days to about 21 days.
The differentiation of hematopoietic cells into NK cells can be evaluated by detecting markers, for example, CD56, CD94, CD117, NKG2D, DNAM-1 and NKp46, for example, by flow cytometry. Differentiation can also be assessed by the morphological characteristics of NK cells, for example, large size, high protein synthesis activity in the abundant endoplasmic reticulum (ER) and / or preformed granules. Maturation of NK cells (e.g., TSNK cells) can be assessed by detecting one or more functionally relevant markers, eg, CD94, CD161, NKp44, DNAM-1, 2B4, NKp46, CD94, KIR, and the NKG2 family of activating receptors (for example, NKG2D). The maturation of NK cells (e.g., TSNK cells) can also be assessed by detecting specific markers during different stages of development. For example, in one embodiment, the pro-NK cells are CD34 +, CD45RA \ CD10 +, CD117 + and / or CD161. "In another embodiment, the immature cells are CD34 +, CD45RA \ CD10", CD117 + and / or CD161" In another embodiment, the immature NK cells are CD34-, CD117 \ CD161 \ NKp46"and / or CD94 / N KG2A. In another embodiment, the CD56brNlante NK cells are CD117 +, NKp46 +, CD94 / NKG2A +, CD16"and / or KIR + '\ In another embodiment, the CD56 cells, where NK are CD117", NKp46 +, CD94 / N KG2 A + / ", CD16 + and / or KIR +.
In a specific embodiment, the maturation of NK cells (e.g., TSNK cells) is determined by the percentage of NK cells (e.g., TSNK cells) which are CD161", CD94 + and / or NKp46 \ In a more specific embodiment, by at least 10%, 20%, 25%, 30%, 35%, 40%, 50%, 55%, 60%, 65% or 70% of mature NK cells (e.g., TSNK cells) are NKp46 +. other more specific modalities, at least 10%, 20%, 25%, 30%, 35%, 40%, 45% or 50% of mature NK cells (e.g., TSNK cells) are CD94 +. specific, at least 10%, 20%, 25%, 30%, 35%, 40%, 45% or 50% of mature NK cells (e.g., TSNK cells) are CD161. " In certain embodiments, the differentiation of hematopoietic cells into NK cells is evaluated by detecting the level of expression of, for example, CD3, CD7 or CD127, CD10, CD14, CD15, CD16, CD33, CD34, CD56, CD94, CD117, CD161. , NKp44, NKp46, NKG2D, DNAM-1, 2B4 or TO-PRO-3, using, for example, antibodies to one or more of these cellular markers. Such antibodies can be conjugated to a detectable label, for example, as a fluorescent label, for example, FITC, R-PE, PerCP, PerCP-Cy5.5, APC, APC-Cy7 or APC-H7. 6. 3. Isolation of TSNK Cells Methods of isolating natural killer cells are known in the art and can be used to isolate TSNK cells. Natural killer cells can be isolated or enriched by staining cells from a tissue source, for example, peripheral blood, with antibodies to CD56 and CD3, and selecting for CD56 + CD3 cells. "TSNK cells can be isolated using commercially available equipment. for example, the NK cell isolation kit (Miltenyi Biotec) .The TSNK cells can also be isolated or enriched by removing different cells from the NK cells in a population of cells comprising the TSNK cells. TSNK can be isolated or enriched by omission of cells displaying non-NK cell markers, eg, antibodies to one or more of CD3, CD4, CD14, CD19, CD20, CD36, CD66, CD123, H LA DR and / or CD235a (Glycophorin A) Negative isolation can be carried out using commercially available equipment, for example, the NK cell negative isolation equipment (Dynal Biotech) .The cells isolated by these methods can be isolated. n additionally classify, for example, to separate CD16 + and CD16 cells ".
Cell separation can be achieved, for example, by flow cytometry, fluorescence activated cell sorting (FACS), or, preferably, magnetic cell sorting using conjugated microspheres with specific antibodies. The cells can be isolated, for example, using a magnetic activated classification (MACS) technique, a method for separating particles based on their ability to join magnetic spheres (e.g., about 0.5-100 μ? T? Diameter) comprising one or more specific antibodies, for example, anti-CD56 antibodies. Magnetic cell separation can be performed and automated using, for example, an AUTOMACS ™ separator (Miltenyi). A variety of useful modifications can be made to the magnetic microspheres, including covalent addition of antibody that specifically recognizes a particular cell surface molecule or hapten. The spheres are then mixed with the cells to allow binding. The cells then pass through a magnetic field to separate cells having the specific cell surface marker. In one embodiment, these cells can then be isolated and re-mixed with magnetic beads coupled to an antibody against additional cell surface markers. The cells then pass once more through a magnetic field, isolating cells that bind both antibodies. Said cells can then be diluted in separate plates, such as microtiter plates for clonal isolation. 6. 4. Placenta perfusate TSNK cells from hematopoietic cells, e.g., stem or hematopoietic progenitors can be produced from any source, e.g., placental tissue, placental perfusate, umbilical cord blood, placental blood, peripheral blood, spleen, liver, or the like. In certain embodiments, hematopoietic stem cells are combined hematopoietic stem cells from placental perfusate and cord blood from the same placenta used to generalize placental perfusate. The placental perfusate comprising placental perfusate cells obtainable, for example, by the methods described in the U.S. Patents. Nos. 7,045,148 and 7,468,276, the descriptions of which are incorporated herein by reference in their totals. 6. 4.1. Composition of Cell Collection Peripheral perfusate and perfusate cells, from which stem cells or hematopoietic progenitors can be isolated, or useful in tumor suppression or treatment of an individual having tumor cells, cancer or a viral infection, for example, in combination with TSNK cells, as provided herein, can be harvested by perfusion from a mammal, e.g., human postpartum placenta using a placental cell harvesting composition. Perfusate of the placenta can be collected by perfusion of the placenta with any physiologically acceptable solution, for example, a saline solution, culture medium, or a more complex cellular collection composition. A cellular harvesting composition suitable for perfusing a placenta, and for the collection and storage of perfusate cells is described in detail in the application publication of E.U.A. No. 2007/0190042, which is incorporated herein by reference in its entirety.
The cell harvesting composition may comprise any physiologically acceptable solution suitable for the collection and / or culture of stem cells, for example, a saline solution (eg, phosphate buffered saline, Kreb's solution, modified Kreb's solution, Eagle, 0.9% NaCl, etc.), a culture medium (e.g., DMEM, H.DMEM, etc.), and the like.
The cellular harvesting composition may comprise one or more components that tend to retain placental cells, ie, prevent placental cells from drying, or delay the death of placental cells, reduce the number of placental cells in a population of cells that die, or the like, from the moment of harvesting to the moment of cultivation. Such components may be, for example, an apoptosis inhibitor (eg, a caspase inhibitor or JNK inhibitor); a vasodilator (eg, magnesium sulfate, an anti-hypertensive drug, atrial natriuretic peptide (ANP), adrenocorticotropin, corticotropin-releasing hormone, sodium nitroprusside, hydralazine, adenosine triphosphate, adenosine, indomethacin, or magnesium sulfate, an inhibitor of phosphod iesterase, etc.); a necrosis inhibitor (e.g., 2- (1 H-indol-3-yl) -3-pentylamino-maleimide, pyrrolidine dithiocarbamate, or clonazepam); a TNF-a inhibitor; and / or an oxygen carrying perfluorocarbon (eg, perfluorooctyl bromide, perfluorodecyl bromide, etc.).
The cell harvesting composition may comprise one or more tissue degrading enzymes, for example, a metalloprotease, a serine protease, a neutral protease, a hyaluronidase, a RNase, or a DNase, or the like. Such enzymes include, but are not limited to, collagenases (for example, collagenase I, II, III or IV, a collagenase from Clostridium histolyticum, etc.); dispase, thermolysin, elastase, trypsin, LIBERASE, hyaluronidase, and the like.
The cellular harvesting composition may comprise a bacteriocidally or bacteriostatically effective amount of an antibiotic. In certain non-limiting embodiments, the antibiotic is a macrolide (e.g., tobramycin), a cephalosporin (e.g., cephalexin, cephradine, cefuroxime, cefprozil, cefaclor, cefixime, or cefadroxil), a clarithromycin, an erythromycin, a penicillin (e.g. , penicillin V) or a quinolone (for example, ofloxacin, ciprofloxacin or norf loxacin), a tetracycline, a streptomycin, etc. In a particular embodiment, the antibiotic is active against Gram (+) and / or Gram (-) bacteria, for example, Pseudomonas aeruginosa, Staphylococcus aureus, and the like.
The cell harvesting composition may also comprise one or more of the following compounds: adenosine (from about 1 mM to about 50 mM); D-glucose (from about 20 mM to about 100 mM); Magnesium ions (from about 1 mM to about 50 mM); a macromolecule of molecular weight greater than 20,000 daltons, in one embodiment, present in an amount sufficient to maintain endothelial integrity and cell viability (e.g., a colloid that occurs synthetically or naturally, a polysaccharide such as dextran or a polyethylene glycol present in about 25 g / l to about 100 g / l, or from about 40 g / l to about 60 g / l); an antioxidant (for example, butylated hydroxyanisole, butylated hydroxytoluene, glutathione, vitamin C or vitamin E present from about 0.1 M to about 5 mM); an agent that prevents entry of calcium into cells (for example, verapamil present from about 2 μ? to about 25 μ?); nitroglycerin (for example, from about 0.05 g / L to about 0.2 g / L); an anticoagulant, in one embodiment, present in an amount sufficient to help prevent coagulation of residual blood (eg, heparin or hirudin present at a concentration of about 1000 units / I to approximately 100,000 units / l); or a compound containing amiloride (for example, amiloride, ethyl isopropyl amiloride, hexamethylene amiloride, dimethyl amiloride or isobutyl amiloride present from about 1.0 μ to about 5 μ). 6.4.2. Collection and Management of Placenta In general, a human placenta is recovered shortly after its expulsion after birth. In a preferred embodiment, the placenta is retrieved from a patient after informed consent and after taking a complete medical history of the patient and is associated with the placenta. Preferably, the medical history continues after delivery.
Before recovering the perfusate, umbilical cord blood and placental blood are eliminated. In certain modalities, after delivery, the cord blood in the placenta is recovered. The placenta can be subjected to a conventional cord blood recovery process. Typically a needle or cannula is used, with the aid of gravity, to exsanguinate the placenta (see, for example, Anderson, U.S. Patent No. 5,372,581, Hessel et al., U.S. Patent No. 5,415,665). The needle or cannula is usually placed in the umbilical vein and the placenta can be gently massaged to help drain cord blood from the placenta. Said cord blood recovery can be performed commercially, for example, LifeBank, Inc., Cedar Knolls, N.J., ViaCord, Cord Blood Registry and CryoCell. Preferably, the placenta is drained by gravity without further manipulation to minimize tissue disruption during cord blood recovery.
Typically, a placenta is transported from the delivery room or delivery to another location, for example, a laboratory, for cord blood recovery and perfusate collection. The placenta is preferably transported in a sterile, thermally insulated transport device (maintaining the temperature of the placenta between 20-28 ° C), for example, by placing the placenta, with a nearby umbilical cord attached, in a zip-lock plastic bag sterile, which is then placed in an insulated container. In another embodiment, the placenta is transported in a cord blood collection equipment substantially as described in the U.S. patent. No. 7,147,626. Preferably, the placenta is delivered to the laboratory four to twenty-four hours after delivery. In certain embodiments, the proximal umbilical cord is fastened, preferably within 4-5 cm (centimeter) of the placental disc insertion prior to retrieval of cord blood. In other modalities, the proximal umbilical cord is held after retrieval of cord blood but before further processing of the placenta.
The placenta, before collection of the perfusate, can be stored under sterile conditions and at room temperature or at a temperature of 5 to 25 ° C (centigrade). The placenta can be stored for a longer period of forty-eight hours, and preferably for a period of four to twenty-four hours before perfusing the placenta to remove any residual cord blood. The placenta is preferably stored in an anticoagulant solution at a temperature of 5 ° C to 25 ° C (Centigrade). Suitable anticoagulant solutions are well known in the art. For example, a solution of heparin or sodium warfarin may be used. In a preferred embodiment, the anticoagulant solution comprises a heparin solution (eg, 1% w / w in 1: 1000 solution). The exsanguinated placenta is preferably stored for no more than 36 hours before collecting placental perfusate. 6. 4.3. Placenta perfusion Methods of perfusing mammalian placentas and obtaining placental perfusate are described, for example, in Hariri, U.S. Patents. Nos. 7,045,148 and 7,255,879, and in the application publications of E.U.A. Nos. 2007/0190042 and 20070275362, the descriptions of which are incorporated herein by reference in their entireties.
Perfusate solution can be obtained perfused by, for example, saline solution, culture medium or cell harvesting compositions described above, via placental vasculature. In one embodiment, a mammalian placenta is perfused by passage of perfusion solution through one or both of the umbilical artery and umbilical vein. The flow of perfusion solution through the placenta can be achieved using, for example, gravity flow in the placenta.
Preferably, the perfusion solution is forced through the placenta using a pump, for example, a peristaltic pump. The umbilical vein, for example, can be cannulated with a cannula, for example, a TEFLON® or plastic cannula, which is connected to a sterile connecting apparatus, such as a sterile tube. The sterile connecting apparatus is connected to a perfusion manifold.
In preparation for perfusion, the placenta is preferably oriented in such a way that the umbilical artery and umbilical vein are located at the highest point of the placenta. The placenta can be perfused by a perfusion solution through the placental vasculature, or through the vasculature of the placenta and surrounding tissue. In one embodiment, the umbilical artery and the umbilical vein are simultaneously connected to a pipette that is connected via a flexible connector to a reservoir of the perfusion solution. The perfusion solution passes into the vein and umbilical artery. The perfusion solution exudes from and / or passes through the walls of the blood vessels from the surface of the placenta that was fixed to the mother's uterus during pregnancy. The perfusion solution can also be introduced through the opening of the umbilical cord and allowed to flow or percolate out of the openings in the wall of the placenta which are in interface with the maternal uterine wall. In another modality, the perfusion solution is passed through the umbilical veins and collected from the umbilical artery, or passed through the umbilical artery and collected from the umbilical veins, that is, passed through only the placental vasculature (fetal tissue).
In one embodiment, for example, the umbilical artery and the umbilical vein are connected simultaneously, for example, to a pipette that is connected via a flexible connector to a reservoir of the perfusion solution. The perfusion solution is passed into the umbilical vein and artery. The perfusion solution exudes from and / or passes through the walls of the blood vessels in the surrounding tissues of the placenta, and is collected in a suitable open container from the surface of the placenta that was fixed to the mother's uterus during gestation The perfusion solution can also be introduced through the umbilical cord opening and allowed to flow or percolate out of the openings in the wall of the placenta which are in interface with the maternal uterine wall. Placental cells that are harvested by this method, can be referred to as a "pan" method, are typically in a mixture of fetal and maternal cells.
In another modality, the perfusion solution is passed through the umbilical veins and collected from the umbilical artery, or passed through the umbilical artery and collected from the umbilical veins. Placental cells collected by this method, which can be referred to as a "closed circuit" method, are typically almost exclusively fetal.
The closed circuit perfusion method, in one modality, can be performed in the following manner. A postpartum placenta is obtained within approximately 48 hours after birth. The umbilical cord is held and cut above the forceps. The umbilical cord can be discarded, or it can be processed to retrieve, for example, umbilical cord stem cells, and / or to process the umbilical cord membrane for the production of a biomaterial. The amniotic membrane can be retained during perfusion, or it can be separated from the chorion, for example, using direct dissection with the fingers. If the amniotic membrane is separated from the chorion before perfusion, it can, for example, be discarded or processed, for example, to obtain stem cells by enzymatic digestion, or to produce, for example, an amniotic membrane biomaterial, for example, the biomaterial described in the US application publication No. 2004/0048796. After cleaning the placenta of all visible blood clots and residual blood, for example, using sterile gauze, the umbilical cord vessels are exposed, for example, by partially cutting the umbilical cord membrane to expose a cord cross section. The vessels are identified, and open, for example, by advancing a closed crocodile clip across the cut end of each vessel. The apparatus, for example, plastic tube connected to an infusion device or peristaltic Itica pump, is then inserted into each of the placental arteries. The pump can be any pump suitable for the purpose, for example, a peristaltic pump. The plastic tube, connected to a sterile collection container, for example, a blood bag such as a 250 ml collection bag, is then inserted into the placenta vein. Alternatively, the tube connected to the pump is inserted into the placenta vein, and tubes to a collection reservoir (s) are inserted into one or both of the placental arteries. The placenta is then perfused with a volume of perfusion solution, for example, approximately 750 ml of perfusion solution. Cells are then collected in the perfusate, for example, by centrifugation.
In one embodiment, the proximal umbilical cord is held during the perfusion, and more preferably, is held within 4-5 cm (centimeters) of the insertion of the cord in the placental disc.
The first collection of perfusion fluid from a mammalian placenta during the exsanguination process is usually colored with residual red blood cells from cord blood and / or placental blood. The perfusion fluid becomes more colorless as the perfusion proceeds and the residual bead globules are washed out of the placenta. In general, 30 to 100 mL of perfusion fluid is adequate to wash blood from the placenta at the beginning, but more or less perfusion fluid can be used depending on the results observed.
The volume of perfusion fluid used to perfuse the placenta may vary depending on the number of placental cells to be harvested, the size of the placenta, the number of collections to be made from a single placenta, etc. In various embodiments, the volume of perfusion fluid may be from 50 mL to 5000 mL, 50 mL to 4000 mL, 50 mL to 3000 mL, 100 mL to 2000 mL, 250 mL to 2000 mL, 500 mL to 2000 mL, or 750 mL to 2000 mL. Typically, the placenta is perfused with 700-800 mL of perfusion fluid after exsanguination.
The placenta can be perfused a plurality of times over the course of several hours or several days. When the placenta is to be perfused a plurality of times, it can be maintained or cultured under aseptic conditions in a container or other suitable container, and perfused with a cellular harvesting composition, or a standard perfusion solution (e.g., saline solution). normal such as phosphate buffered saline ("PBS") with or without an anticoagulant (eg, heparin, sodium warfarin, coumarin, bíshydroxycoumarin) and / or with or without an antimicrobial agent (eg, β-mercaptoethanol (0.1 mM), antibiotics such as streptomycin (for example, 40-100 μg ml), penicillin (for example, at 40U / ml), amphotericin B (for example, at 0.5μ? / ???). Isolated placenta is maintained or cultured for a period of time without collecting the perfusate, so that the placenta is maintained or cultured for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours, or 2 or 3 0 more days before perfusion and perfusate collection. The perfused placenta can be maintained for one or more additional time (s), for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or more hours, and perfused a second time with, for example, 700-800 mL perfusion fluid. The Placenta can be perfused 1, 2, 3, 4, 5 or more times, for example, once every 1, 2, 3, 4, 5 or 6 hours. In a preferred embodiment, perfusion of the placenta and collection of perfusion solution, eg, placental cell harvesting composition, is repeated until the number of nucleated cells recovered falls below 100 cells / ml. Perfusates at different time points can be processed more individually to recover cell-dependent populations of time, e.g., total nucleated cells. You can also group perfused different time points. 6. 4.4. Perfusate of Placenta and Cells Placenta Perfusate Typically, the placental perfusate from a single placental perfusion comprises from about 100 million to about 500 million nucleated cells, including hematopoietic cells from which TSNK cells can be produced by the method described herein. In certain embodiments, the placental cells or perfusate cells comprise CD34 + cells, e.g., stem cells or hematopoietic progenitors. Said cells can, in a more specific embodiment, comprise CD34 + CD45"progenitor or stem cells, CD34 + CD45 + stem or progenitor cells, or the like In certain embodiments, the perfusate or perfusate cells are cryopreserved before the isolation of hematopoietic cells. In certain other embodiments, the placental perfusate comprises, or the perfusate cells comprise, only fetal cells, or a combination of fetal cells and maternal cells. 6. 5. TSNK cells In one aspect, TSNK cells are provided herein, the NK cells produced by the methods described herein (eg, two-step method). Furthermore, a population of cells comprising the TSNK cells produced by the methods described herein (eg, two-step method) is provided herein. In a specific embodiment, said NK cells (e.g., TSNK cells) are CD3"CD56 + CD16". In another specific embodiment, said NK cells (e.g., TSNK cells) are additionally CD94 + CD117 +. In another specific embodiment, said NK cells (e.g., TSNK cells) are additionally CD161. "In another specific embodiment, said NK cells (e.g., TSNK cells) are additionally NKG2D +. In another specific embodiment, said NK cells are additionally NKp46 + In another specific embodiment, said NK cells are additionally CD226 +.
In certain modalities, more than 50%, 60%, 70%, 80%, 90%, 92%, 94%, 96%, 98% of said TSNK cells are CD56 + and CD16 \ In other modalities, at least 50% , 60%, 70%, 80%, 82%, 84%, 86%, 88% or 90% of said TSNK cells are CD3"and CD56 + In other embodiments, at least 50%, 52%, 54%, 56%, 58% or 60% of said TSNK cells are NKG2D In other modalities, less than 30%, 20%, 10%, 9%, 8%, 7%, 6%, 5%, 4% or 3% of said cells are NKB1 + In certain other modalities, less 30%, 20%, 10%, 8%, 6%, 4% or 2% of said TSNK cells are NKAT2 + In certain other modalities, less than 30%, 20%, 10%, 8%, 6% , 4% or 2% of said TSNK cells are CD56"and CD16 +. In more specific modalities, at least 10%, 20%, 25%, 30%, 35%, 40%, 50%, 55%, 60%, 65% or 70% of said TSNK CD3 \ CD56 + cells are NKp46 + . In other more specific modalities, at least 10%, 20%, 25%, 30%, 35%, 40%, 50%, 55%, 60%, 65%, 70%, 75%, 80% or 85% of said TSNK CD3", CD56 + cells are CD117 \ In other more specific modalities, at least 10%, 20%, 25%, 30%, 35%, 40%, 45% or 50% of said TSNK CD3 cells", CD56 + are CD94 +. In other more specific embodiments, at least 10%, 20%, 25%, 30%, 35%, 40%, 45% or 50% of said TSNK CD3", CD56 * cells are CD161". In other more specific modalities, at least 10%, 12%, 14%, 16%, 18% or 20% of said TSNK CD3"cells, CD56 + are CD226 +, in more specific modalities, at least 20%, 25% , 30%, 35% or 40% of said TSNK CD3 \ CD56 + cells are CD7 + In more specific modalities, at least 30%, 35%, 40%, 45%, 50%, 55% or 60% of said cells TSNK CD3", CD56 + are CD5 +.
In several other embodiments, TSNK cells can be combined with, for example, NK cells, wherein said NK cells have been isolated from a tissue source and have not expanded; NK cells isolated from a tissue source and expanded, or NK cells produced by a different method, for example, natural killer cells CD56 + CD16 +, for example, in ratios of, for example, about 1:10, 2: 9 3: 8, 4: 7, 5: 6, 6: 5, 7: 4, 8: 3, 9: 2, 1:10, 1: 9, 1: 8, 1: 7, 1: 6, 1 : 5, 1: 4, 1: 3, 1: 2, 1: 1, 2: 1, 3: 1, 4: 1, 5: 1, 6: 1, 7: 1, 8: 1 or approximately 9: 1. As used in this context, "isolated" means that the cells have been removed from their normal tissue environment.
TSNK cells may have a fetal genotype or a maternal genotype. For example, since the postpartum placenta, as a source of suitable hematopoietic cells to produce TSNK cells, comprises tissue and cells from the fetus and from the mother, the placental perfusate may comprise only fetal cells, or a substantial majority of cells fetal (eg, more than about 90%, 95%, 98% or 99%) or may comprise a mixture of fetal and maternal cells (eg, fetal cells comprise less than about 90%, 80%, 70%, 60% or 50% of the total nucleated cells of the perfusate). In one embodiment, TSNK cells are derived only from hematopoietic cells from fetal placenta, eg, cells obtained from closed-circuit perfusion of the placenta wherein the perfusate produces perfusate comprising a substantial majority, or only, hematopoietic cells of fetal placenta. In another embodiment, TSNK cells are derived from fetal and maternal cells, for example, cells obtained by perfusion by the pan method (see above), wherein perfusion produced perfusate comprising a mixture of fetal and maternal placental cells. Thus, in one embodiment, a population of intermediate natural killer cells derived from the placenta, the substantial majority of which have the fetal genotype, is provided herein. In another embodiment, a population of intermediate natural killer cells derived from placenta are provided which comprise natural killer cells that have the fetal genotype and natural killer cells that have the maternal phenotype.
Also provided herein are populations of TSNK cells comprising natural killer cells not produced by the methods described herein. For example, in one embodiment, a population of TSNK cells that also comprises natural killer cells isolated from, for example, umbilical cord blood, peripheral blood, bone marrow, or a combination of two or more of the above is provided. , or NK cells expanded by a method different from the methods described herein. Said populations of TSNK cells may comprise TSNK cells and other NK cells in, for example, a ratio of approximately 1:10, 2: 9, 3: 8, 4: 7, 5: 6, 6: 5, 7: 4 8: 3, 9: 2, 10: 1, 1: 9, 1: 8, 1: 7, 1: 6, 1: 5, 1: 4, 1: 3, 1: 2, 1: 1, 2 : 1, 3: 1, 4: 1, 5: 1, 6: 1, 7: 1, 8: 1, 9: 1, 100: 1, 95: 5, 90:10, 85:15, 80:20 , 75:25, 70:30, 65:35, 60:40, 55:45, 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15 : 85, 10:90, 5:95, 100: 1, 95: 1, 90: 1, 85: 1, 80: 1, 75: 1, 70: 1, 65: 1, 60: 1, 55: 1 50: 1, 45: 1, 40: 1, 35: 1, 30: 1, 25: 1, 20: 1, 15: 1, 10: 1, 5: 1, 1: 1, 1: 5, 1 : 10, 1:15, 1:20, 1:25, 1:30, 1:35, 1:40, 1:45, 1:50, 1:55, 1:60, 1:65, 1:70 , 1:75, 1:80, 1:85, 1:90, 1:95 or about 1: 100 or similar.
In certain embodiments, isolated natural killer cells (e.g., TSNK cells) or populations enriched for natural killer cells (e.g., TSNK cells) can be evaluated by detecting one or more functionally relevant markers, e.g., CD94, CD161, NKp44, DNAM-1, 2B4, NKp46, CD94, KIR and the NKG2 family of activating receptors (e.g., NKG2D). In some embodiments, the purity of isolated or enriched natural killer cells can be confirmed by detecting one or more of CD56, CD3 and CD16.
Optionally, the cytotoxic activity of isolated or enriched natural killer cells can be evaluated, for example, in a cytotoxicity assay using tumor cells, for example, cultured tumor cells K562, LN-18, U937, WER1-RB-1, U- 118MG, HT-29, HCC2218, KG-1 or U266, or the like as target cells. 6. 6. TSNK Cells in Combination with Placental Perfusate Also herein are provided compositions comprising TSNK cells in combination with placental perfusate, placental perfusate cells and / or adherent placental cells, for example, for use in suppressing the proliferation of a tumor cell or plurality of tumor cells. 6. 6.1. Combinations of TSNK and Perfused Cells or Perfusion Cells Also herein are provided compositions comprising combinations of TSNK cells and placental perfusate and / or placental perfusate cells. In one embodiment, for example, a placental perfusate volume supplemented with TSNK cells is provided herein. In specific embodiments, for example, each milliliter of placental perfusate is supplemented with approximately 1 x 104, 5 x 104, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108 or more TSNK cells. In another embodiment, placental perfusate cells are supplemented with TSNK cells. In certain other embodiments, when placental perfusate cells are combined with TSNK cells, the placental perfusate cells in general comprise about, more than about, or less than about, 50%, 45%, 40%, 35%, %, 25%, 20%, 15%, 10%, 8%, 6%, 4%, 2% or 1% of the total number of cells. In certain other embodiments, when TSNK cells are combined with a plurality of placental perfusate cells and / or combined natural killer cells, the NK cells in general comprise about, more than about, or less than about, 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, 8%, 6%, 4%, 2% or 1% of the total number of cells. In certain other embodiments, when TSNK cells are used to supplement placental perfusate, the volume of solution (eg, saline, culture medium or the like) in which the cells are suspended comprises about, more than about, or less than about , 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, 8%, 6%, 4%, 2% or 1% of the total volume of perfusate plus cells, wherein the TSNK cells are suspended at about 1 x 104, 5 x 10 x 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108 or more cells per milliliter before supplementation.
In other embodiments, any of the above combinations of cells, in turn, is combined with umbilical cord blood or umbilical cord blood nucleated cells.
In addition, placenta perfusate is obtained here which is obtained from two or more sources, for example, two or more placentas, and are combined, for example, grouped. Said pooled perfusate may comprise approximately equal volumes of perfusate from each source, or may comprise volumes different from each source. The relative volumes of each source can be selected randomly, or based on, for example, a concentration or amount of one or more cellular factors, eg, cytokines, growth factors, hormones or the like; the number of placental cells perfused from each source; or other perfused characteristics of each source. The perfusate of multiple infusions of the same placenta can be grouped similarly.
Similarly, placental perfusate cells, and intermediate natural killer cells derived from placenta are provided herein, which are obtained from two or more sources, eg, two or more placentas, or are pooled. Said pooled cells may comprise approximately equal numbers of cells from two or more sources, or different numbers of cells from one or more of the pooled sources. The relative numbers of cells of each source can be selected based on, for example, the number of one or more specific cell types in the cells to be grouped, for example, the number of CD34 + cells, etc.
Also herein are provided TSNK cells, and combos of TSNK cells with placental perfusate and / or placental perfusate cells, which have been tested to determine the extent or amount of tumor suppression (i.e., potency) to be expected from, for example, a given number of TSNK cells, or a given volume of perfusate. For example, an aliquot or cell sample number is contacted with a known number of tumor cells under conditions where the tumor cells would otherwise proliferate, and the proliferation rate of the tumor cells in the presence of placental perfusate, cells of perfusate, and the proliferation rate of tumor cells in the presence of perfusate of placenta, perfusate cells, natural killer cells of placenta, or combinations thereof, over time (eg, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 weeks, or more) is compared to the proliferation of an equivalent number of tumor cells in the absence of perfusate, perfusate cells, natural killer cells of placenta, or combinations thereof. The potency of the cells can be expressed, for example, as the number of cells or volume of solution required to suppress tumor cell growth, for example, by approximately 10%, 15%, 20%, 25%, 30%, 35% , 40%, 45%, 50%, or similar.
In certain modalities, TSNK cells are provided as administrable units of pharmaceutical grade. Said units can be supplied in discrete volumes, for example, 15 ml_, 20 ml_, 25 ml, 30 ml, 35 ml_, 40 ml, 45 ml_, 50 ml_, 55 ml_, 60 ml_, 65 ml_, 70 ml, 75 ml , 80 mL, 85 mL, 90 mL, 95 mL, 100 mL, 150 mL, 200 mL, 250 mL, 300 mL, 350 mL, 400 mL, 450 mL, 500 mL, or the like. Such units can be provided to contain a specified number of cells, for example, TSNK cells alone, or TSNK cells in combination with other NK cells or perfusate cells, for example, 1 x 104, 5 x 104, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108 or more cells per milliliter, or 1 x 104, 5 x 104, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108, 1 x 109, 5 x 109, 1 x 1010, 5 x 1010, 1 x 1011 or more cells per unit. In specific embodiments, the units may comprise approximately, at least approximately, or at most approximately 1 x 104, 5 x 104, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107 , 1 x 108, 5 x 108 or more cells per milliliter, or 1 x 104, 5 x 104, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108 , 5 x 108, 1 x 109, 5 x 109, 1 x 1010, 5 x 1010, 1 x 1011 or more cells per unit. Said units may be provided to contain specified numbers of TSNK cells, and / or any other cell.
In the above embodiments, TSNK cells or combinations of TSNK cells with other NK cells, perfusate or perfusate cells can be autologous to a recipient (i.e., obtained from the container), or allogeneic to a recipient (i.e., obtained from at least one other individual from said vessel).
In certain embodiments, each unit of cells is marked to specify one or more of volume, number of cells, type of cells, whether the unit has been enriched by a particular type of cell, and / or power of a given number of cells. cells in a unit, or a given number of milliliters of the unit, that is, if the cells in the unit cause a measurable suppression of proliferation of a particular type or types of tumor cell. 6. 6.2. Combinations of TSNK Cells and Cells Adherent Placenta Mother In other embodiments, TSNK cells, either alone or in combination with placental perfusate or placental perfusate cells, are supplemented with isolated adherent placental cells, e.g., placental stem cells and multipotent placental cells as described, for example, in the US patents Nos. De Hariri 7,045,148 and 7,255,879, and in the patent application publication of E.U.A. Not 2007/0275362, the descriptions of which are incorporated herein by reference in their totals. "Adherent placental cells" means that the cells are adherent to a tissue culture surface, eg, tissue culture plastic. Adherent placental cells useful in the compositions and methods described herein are not trophoblasts, embryonic germ cells or embryonic stem cells. In certain embodiments, adherent placental stem cells are used as feeder cells during the processes (eg, two-step method) as described above.
TSNK cells, either alone or in combination with placental perfusate or placental perfusate cells can be supplemented with, for example, 1 x 104, 5 x 104, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108 or more cells per milliliter, or 1 x 104, 5 x 104, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5? 108, or more adherent placental cells per milliliter, or 1 x 10 5 x 1 O 4, 1 x 105, 5 x 105, 1 x 1 O 6, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108 or more cells per milliliter, or 1 x 104, 5 x 104, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108, 1 x 109, 5 x 109, 1 x 1010, 5 x 1010, 1 x 1011 or more adherent placental cells. Adherent placental cells in the combinations can be, for example, adherent placental cells that have been cultured for, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14 , 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38 or 40 population doublings, or more.
Isolated adherent placental cells, when cultured in primary cultures or expanded in cell culture, adhere to the tissue culture substrate, eg, tissue culture container surface (eg, tissue culture plastic). The adherent placental cells in culture assume a starry appearance usually fibroblast, with a number of cytoplasmic processes that extend from the central cell body. However, adherent placental cells morphologically distinguishable from fibroblasts cultured under the same conditions, such as adherent placental cells exhibit a greater number of such processes than fibroblasts. Morphologically, the adherent placental cells are also distinguishable from hematopoietic stem cells, which usually assume a more rounded, or cobbled, morphology in culture.
Isolated adherent placental cells, and adherent placental cell populations, useful in the compositions and methods provided herein, express a plurality of markers that can be used to identify and / or isolate cells, or populations of cells comprising adherent placental cells. Adherent placental cells, and adherent placental cell populations useful in the compositions and methods provided herein include adherent placental cells and cell populations containing adherent placental cells obtained directly from the placenta, or any part thereof ( for example, amnion, chorion, amnion-chorion plaque, placenta cotyledons, umbilical cord, and the like). The adherent placental stem cell population, in one embodiment, is a population (i.e., two or more) of adherent placental stem cells in culture, eg, a population in a container, e.g., a bag.
Adherent placental cells generally express the markers CD73, CD105, and CD200, and / or OCT-4, and do not express CD34, CD38 or CD45. Adherent placental stem cells can also express HLA-ABC (MHC-1) and HLA-DR. These markers can be used to identify adherent placental cells, and to distinguish adherent placental cells from other cell types. Since adherent placental cells can express CD73 and CD105, they can have mesenchymal stem cell type characteristics. The lack of expression of CD34, CD38 and / or CD45 identifies adherent placental stem cells as non-hematopoietic stem cells.
In certain embodiments, the isolated adherent placental cells described herein suppress detectably cancer cell proliferation or tumor growth.
In certain embodiments, the isolated adherent placental cells are isolated placental stem cells. In certain other embodiments, isolated adherent placental cells are multipotent isolated placental cells. In a specific embodiment, the isolated adherent placental cells are CD34", CD10 + and CD105 + as detected by flow cytometry In a more specific embodiment, isolated adherent placental cells CD34", CD1CT, CD105 + are placental stem cells . In another more specific embodiment, the isolated placental cells CD34", CD10 +, CD105 + are multipotent adherent placental cells.In another specific embodiment, isolated placental cells CD34", CD10 +, CD105 + have the potential to differentiate into cells of a phenotype neutral, cells of an osteogenic phenotype, or cells of a chondrogenic phenotype. In a more specific embodiment, the adherent placenta cells isolated CD34", CD10 +, CD105 * are additionally CD200 + In another more specific embodiment, the isolated adherent placenta cells CD34", CD10 +, CD105 + are additionally CD90 + or CD45" , as detected by flow cytometry In another more specific embodiment, isolated adherent placenta cells CD34", CD10 +, CD105 + are additionally CD90 + or CD45", as detected by flow cytometry. adherent placental cells CD34", CD10 +, CD105 +, CD200 + are additionally CD90 + or CD45", as detected by flow cytometry In another more specific modality, adherent placental cells CD34", CD10 +, CD105 +, CD200 + they are additionally CD90 * and CD45", as detected by flow cytometry. In another more specific embodiment, adherent placental cells CD34", CD10 +, CD105", CD200 \ CD90 +, CD45" are additionally CD80"and CD 86", as detected by flow cytometry.
In one embodiment, isolated adherent placental cells are CD200 +, HLA-G +. In a specific embodiment, said isolated adherent placental cells are also CD73 + and CD105 +. In another specific embodiment, said isolated adherent placental cells are also CD34", CD38" or CD45. "In a more specific embodiment, said isolated adherent placental cells are also CD34", CD38", CD45", CD73 + and CD105 +. In another embodiment, said isolated adherent placental cells produce one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies.
In another embodiment, the isolated adherent placental cells are CD73 +, CD105 +, CD200. In a specific embodiment of said populations, said isolated adherent placental cells are also HLA-G +. In another specific embodiment, said isolated adherent placental cells are also CD34", CD38" or CD45. "In another specific embodiment, said isolated adherent placental cells are also CD34", CD38 'and CD45. "In a more specific embodiment, said isolated adherent placental cells are also CD34", CD38", CD45"and HLA-G \ In another specific embodiment, said isolated adherent placental cells produce one or more embryoid-like bodies when cultured under conditions that allow the formation of bodies. of embryoid type.
In another embodiment, isolated adherent placental cells are CD200 +, OCT-4 +. In a specific embodiment, said isolated adherent placental cells are also CD73 + and CD105 +. In another specific embodiment, said isolated adherent placental cells are also HLA-G +. In another specific embodiment, said isolated adherent placental cells are also CD34 ', CD38"and CD45". In a more specific embodiment, said isolated adherent placental cells are also CD34", CD38", CD45", CD73" CD105 + and HLA-G +. "In another specific embodiment, the isolated adherent placental cells also produce one or more bodies of embryoid type when grown under conditions that allow the formation of embryoid-like bodies.
In another embodiment, isolated adherent placental cells are CD73 +, CD105 + and HLA-G +. In a specific embodiment, isolated adherent placental cells are also CD34", CD38" or CD45. "In another specific embodiment, said isolated adherent stem cells are also OCT-4 + In another specific embodiment, said adherent stem cells are also CD200 * In a modality more specific, said adherent stem cells are also CD34", CD38", CD45 \ OCT-4 + and CD200 +.
In another embodiment, isolated adherent placental cells are CD73 +, CD105 + stem cells, wherein said cells produce one or more embryoid-like bodies under conditions that allow the formation of embryoid-like bodies. In a specific embodiment, said isolated adherent placental cells are also CD34", CD38" or CD45. "In another specific embodiment, the isolated adherent placental cells are also CD34", CD38"and CD45". In another specific embodiment, isolated adherent placental cells are also OCT-4 +. In a more specific embodiment, said isolated adherent placental cells are also OCT-4 *, CD34", CD38" and CD45 In another embodiment, the isolated adherent placental cells are OCT-4 + stem cells, wherein said isolated adherent placental cells produce one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies, and wherein said stem cells have been identified as detectably suppressing cancer cell proliferation or tumor growth.
In various modalities, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80 %, at least 90% or at least 95% of said isolated adherent placental cells are OCT-4 +. In a specific embodiment of the above populations, said isolated adherent placental cells are also CD73 + and CD105 +. In another specific embodiment, said isolated adherent placental cells are also CD34", CD38" or CD45. "In another specific embodiment, said stem cells are CD200.In a more specific embodiment, said isolated adherent placental cells are also CD73. CD105 +, CD200 +, CD43", CD38" and CD45". In another specific embodiment, said isolated adherent placental cells have expanded, for example, passed at least once, at least three times, at least five times, at least 10 times, at least 15 times or by at least 20 times.
In a more specific embodiment of any of the foregoing embodiments, isolated adherent placental cells express ABC-p (a placental-specific ABC transport protein, see, eg, Allikmets et al., Cancer Res. 58 (23): 5337 -9 (1998)).
In another embodiment, isolated adherent placental cells CD29 \ CD44 \ CD73 \ CD90 +, CD105 +, CD200 +, CD34"and CD133 \ In another embodiment, the isolated adherent placental cells secrete constitutively IL-6, IL-8 and Monocyte chemoattractant protein (MCP-1).
Each of the isolated adherent placental cells referred to above may comprise cells obtained and isolated directly from a mammalian placenta., or cells that have been cultured and passed at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 25, 30 or more times, or a combination of them. The tumor cell suppressor pluralities of the isolated adherent placental cells described above may comprise at least, or not more than, 1 x 104, 5 x 10 4, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108, 1 x 109, 5 x 109, 1 x 1010, 5 x 1010, 1 x 1011 or more 1 x 104, 5 x 104, 1 x 105 , 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108, 1 x 109, 5 x 109, 1 x 1010, 5 x 1010, 1 x 1011 or cells of placenta adherents isolated. 6. 6.3. Compositions that Include Media Conditioning of Adherent Placental Cells The use of a composition comprising TSNK cells and additionally conditioned medium is also provided herein, wherein said composition is tumor suppressor, or is effective in the treatment of cancer or viral infection. Adherent placental cells as described in section 6.6.2, above can be used to produce conditioned media that is tumor cell, anti-cancer or anti-viral cell suppressor, ie, medium comprising one or more secreted biomolecules or excreted by cells that have a detectable tumor cell suppressive effect, anti-cancer effect or antiviral effect. In various embodiments, the conditioned medium comprises medium in which the cells have proliferated (i.e., been cultured) for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or more days. In other embodiments, the conditioned medium comprises medium wherein said cells have grown to at least 30%, 40%, 50%, 60%, 70%, 80%, 90% confluency. Said conditioned medium can be used to support the culture of a separate population of cells, for example, placental cells, or cells of another class. In another embodiment, the conditioned medium provided herein comprises medium wherein the isolated adherent placental cells, for example, isolated adherent placental stem cells or isolated adherent placental multipotent cells, and cells different from the isolated adherent placental cells, for example, non placental stem cells or multipotent cells, have been cultured.
Said conditioned medium can be combined with any of, or any combination of TSNK cells, placental perfused, placental perfusate cells to form a composition that is tumor cell suppressor, anti-cancer or antiviral. In certain embodiments, the composition comprises less than half of conditioned media in volume, eg, about, or less than about, 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15% , 10%, 5%, 4%, 3%, 2% or 1% by volume.
Thus, in one embodiment, a composition comprising TSNK cells and culture medium is provided herein from a culture of isolated adherent placental cells, wherein said isolated adherent placental cells (a) are adhere to a substrate; and (b) are CD34, CD10 + and CD105 +; wherein said composition detectably suppresses the growth or proliferation of tumor cells, or is anti-cancer or antiviral. In a specific embodiment, the isolated adherent placental cells are CD34", CD10 + and CD105 +, as detected by flow cytometry In a more specific embodiment, the isolated adherent placenta cells CD34", CD10 +, CD105 + are stem cells from placenta. In another more specific embodiment, the isolated placental cells CD34 ', CD10 +, CD105 + are multipotent adherent placental cells. In another specific embodiment, the isolated placenta cells CD34 ', CD10 +, C105 + have the potential to differentiate cells of a neural phenotype, cells of an osteogenic phenotype, or cells of a chondrogenic phenotype. In a more specific embodiment, the adherent placenta cells isolated CD34", CD10 +, CD105 + are additionally CD200 + In another more specific embodiment, the adherent placenta cells isolated CD34", CD10 +, CD105 + are additionally CD90 + or CD45"as detected by flow cytometry In another more specific modality, isolated adherent placenta cells CD34", CD10 +, CD105 +, CD200 + are additionally CD90 + or CD45", as detected by flow cytometry. adherent placental cells CD34", CD10 +, CD105 +, CD200 + are additionally CD90 + and CD45", as detected by flow cytometry In another more specific modality, adherent placental cells CD34", CD10 \ CD105 +, CD200 + , CD90 +, CD45"are additionally CD80" and CD86", as detected by flow cytometry.
In another embodiment, a composition comprising TSNK cells and culture medium is provided from a culture of isolated adherent placental cells, wherein said isolated adherent placental cells (a) adhere to a substrate; and (b) express CD200 and HLA-G, or express CD73, CD105 and CD200, or express CD200 and OCT-4, or express CD73, CD105 and HLA-G, or express CD73 and CD105 and facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising the placental stem cells when said population is cultured under conditions that allow the formation of embryoid-like bodies, or express OCT-4 and facilitate the formation of one or more bodies of embryoid type in a population of placental cells comprising the placental stem cells when said population is cultured under conditions that allow the formation of embryoid-like bodies; wherein said composition detectably suppresses the growth or proliferation of tumor cells, or is anti-cancer or antiviral. In a specific embodiment, the composition further comprises a plurality of said isolated adherent placental cells. In another specific embodiment, the composition comprises a plurality of non-placental cells. In a more specific embodiment, said non-placental cells comprise CD34 + cells, for example, hematopoietic progenitor cells, such as peripheral blood hematopoietic progenitor cells, cord blood hematopoietic progenitor cells, or hematopoietic blood progenitor cells of placenta. Non-placental cells can also comprise stem cells, such as mesenchymal stem cells, e.g., mesenchymal stem cells derived from bone marrow. Non-placental cells can also be one or more types of adult cells or cell lines. In another specific embodiment, the composition comprises an antiproliferative agent, for example, an a nti-M I P-1 a or anti-M I P-1β antibody.
In a specific embodiment, the culture medium conditioned by one of the cells or cell combinations described above is obtained from a plurality of isolated adherent placental cells co-cultured with a plurality of tumor cells from about 1: 1 to about 2: 1, approximately 3: 1, approximately 4: 1 or approximately 5: 1 isolated adherent placental cells to tumor cells. For example, the conditioned culture medium or supernatant can be obtained from a culture comprising about 1 × 10 5 isolated adherent placenta cells, about 1 × 10 6 isolated adherent placenta cells, about 1 × 10 7 isolated or approximately adherent placental cells. 1 x 108 adherent placental cells isolated, or more. In another specific embodiment, the conditioned culture medium or supernatant is obtained from a co-culture comprising from about 1 × 10 5 to about 5 × 10 5 isolated adherent placenta cells and about 1 × 10 5 tumor cells; from about 1 x 106 to about 5 x 106 isolated adherent placental cells and about 1 x 10 6 tumor cells; from about 1 x 107 to about 5 x 107 isolated adherent placental cells and about 1 x 107 tumor cells; or from about 1 x 108 to about 5 x 108 isolated adherent placental cells and about 1 x 10 8 tumor cells. 6. 7. Conservation of Cells Cells, e.g., TSNK cells or placental perfusate cells comprising stem cells or hematopoietic progenitor cells, can be preserved, i.e., placed under conditions that allow long-term storage, or under conditions that inhibit cell death, e.g. by apoptosis or necrosis.
Placental perfusate can be produced by passage of a cellular harvesting composition through at least part of the placenta, for example, through the placental vasculature. The cellular harvesting composition comprises one or more compounds that act to preserve cells contained within the perfusate. Said placental cell harvesting composition may comprise an apoptosis inhibitor, necrosis inhibitor and / or an oxygen carrier perfluorocarbide, as described in the application publication of E.U.A. No. 20070190042 related, the description of which is incorporated herein by reference in its entirety.
In one embodiment, the perfusate or a population of placental cells are harvested from a mammalian postpartum placenta, eg, human, by contacting the perfusate or population of cells with a cellular harvesting composition comprising an apoptosis inhibitor and a oxygen carrier perfluorocarbide, wherein said apoptosis inhibitor is present in an amount and for a time sufficient to reduce or prevent apoptosis in the placental cell population, for example, adherent placental cells, for example, placental stem cells or multipotent placental cells, compared to a population of cells not contacted with the apoptosis inhibitor. For example, the placenta can be perfused with the cellular harvesting composition, and placental cells, for example, total nucleated placental cells, are isolated therefrom. In a specific embodiment, the apoptosis inhibitor is a caspase inhibitor. In another specific embodiment, said inhibitor of apoptosis is a JNK inhibitor. In a more specific embodiment, said JNK inhibitor does not modulate the differentiation or proliferation of adherent placental cells, for example, adherent placental stem cells or adipose multipotent placental cells. In another embodiment, the cellular harvesting composition comprises said apoptosis inhibitor and said oxygen carrying perfluorocarbon in separate phases. In another embodiment, the cellular harvesting composition comprises said apoptosis inhibitor and said oxygen carrying perfluorocarbon in an emulsion. In another embodiment, the cellular harvest composition additionally comprises an emulsifier, for example, lecithin. In another embodiment, said apoptosis inhibitor and said perfluorocarbon are between about 0 ° C and about 25 ° C at the time of contacting the placental cells. In another more specific embodiment, said apoptosis inhibitor and said perfluorocarbon are between about 2 ° C and 10 ° C, or between about 2 ° C and about 5 ° C, at the time of contacting the placental cells. In another more specific embodiment, said contact is made during transport of said population of cells. In another more specific embodiment, said contact is made during the freezing and thawing of said population of cells.
In another embodiment, placental perfusate and / or placental perfusate cells can be harvested and preserved upon contacting the perfusate and / or cells with an apoptosis inhibitor and an organ preservative compound, wherein said apoptosis inhibitor is present in a amount and for a sufficient time to reduce or prevent apoptosis of cells, compared to perfused or placental cells not contacted with the apoptosis inhibitor. In a specific embodiment, the oxygen-conserving compound is UW solution (described in U.S. Patent No. 4,798,824, also known as VIASPAN ™, see also Soouthard et al., Transplantation 49 (2): 251-257 (1990) or solution described in Stern et al, U.S. Patent No. 5,552,267, the descriptions of which are incorporated herein by reference in their entireties In another embodiment, said organ-conserving composition is hydroxyethyl starch, lactobionic acid, raffinose or a combination thereof In another embodiment, the cellular harvesting composition of placenta further comprises an oxygen carrying perfluorocarbon, either in two phases or as an emulsion.
In another embodiment of the method, the placental cells are contacted with a cellular harvesting composition comprising an oxygen carrier perfluorocarbon inhibitor, organ preservative compound, or combination thereof, during perfusion. In another embodiment, the placental cells are contacted with said cell harvesting compound after collection by perfusion.
Typically, during collection, enrichment and cellular isolation of placenta, it is preferable to minimize or eliminate cellular stress due to hypoxia and mechanical stress. In another embodiment of the method, therefore, placental perfusate or a population of placental cells is exposed to a hypoxic condition during collection, enrichment or isolation for less than six hours during said storage, wherein a hypoxic condition is a concentration of oxygen that is less than the normal concentration of blood oxygen. In a more specific embodiment, said perfusate or population of placental cells is exposed to said hypoxic condition for less than two hours during said storage. In other more specific embodiments, said population of placental cells is exposed to said hypoxic condition for less than one hour, or less than thirty minutes, or is not exposed to a hypoxic condition, during collection, enrichment or isolation. In another specific embodiment, said population of placental cells is not exposed to shear stress during collection, enrichment or isolation. Cells, for example, placental perfusate cells, hematopoietic cells, e.g., CD34 + hematopoietic stem cells; NK cells, for example, TSNK cells; Isolated adherent placental cells provided herein may be cryopreserved, for example, in cryopreservation medium in small containers, for example, ampoules or septum vials. In certain embodiments, the cells provided herein are cryopreserved at a concentration of approximately 1 x 104-5 x 108 cells per mL. In more specific embodiments, the cells provided herein are cryopreserved at a concentration of about 1 × 10 4, 5 × 10 4, 1 × 10 5, 5 × 10 5, 1 × 10 6, 5 × 10 6, 1 × 107, 5 × 10 7 cells per mL.
Suitable cryopreservation medium includes, but is not limited to, normal saline, culture medium including, for example, growth medium, or cell freezing medium, for example, commercially available cell freezing medium, for example, C2695, C2639 or C6039 (Sigma); CryoStor® CS2, CryoStor® CS5 or CryoStor® CS10 (BioLife Solutions). The cryopreservation medium preferably comprises DMSO (dimethyl sulfoxide), at a concentration of, for example, about 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10% (v / v). The cryopreservation medium may comprise additional agents, for example, methylcellulose, dextran, albumin (for example, human serum albumin), trehalose, and / or glycerol. In certain embodiments, the cryopreservation medium comprises about 1% -10% DMSO, about 25% -75% dextran and / or about 20-60% human serum albumin (HSA). In certain embodiments, the cryopreservation medium comprises about 1% -10% DMSO, about 25% -75% trehalose and / or about 20-60% human HSA. In a specific embodiment, the cryopreservation medium comprises 5% DMSO, 55% dextran and 40% HSA. In a more specific embodiment, the cryopreservation medium comprises 5% DMSO, 55% dextran (10% w / v in normal saline) and 40% HSA. In another specific embodiment, the cryopreservation medium comprises 5% DMSO, 55% trehalose and 40% HSA. In a more specific embodiment, the cryopreservation medium comprises 5% DMSO, 55% trehalose (10% w / v in normal saline) and 40% HSA. In another specific embodiment, the cryopreservation medium comprises CryoStor® CS5. In another specific embodiment, the cryopreservation medium comprises CryoStor® CS10.
The cells provided herein can be cryopreserved by any of a variety of methods, and at no stage of cell culture, expansion or differentiation. For example, the cells provided herein may be cryopreserved immediately after isolation of the tissues or organs of origin, for example, perfused placenta or umbilical cord blood, or during, or after, the first or second step of the delineated methods. before. In certain embodiments, the hematopoietic cells, e.g., hematopoietic stem or progenitor cells are cryopreserved within about 1, 5, 10, 15, 20, 30, 45 minutes or within about 1, 2, 4, 6, 10, 12 , 18, 20 or 24 hours after isolation from the tissues or organs of origin. In certain embodiments, said cells are cryopreserved within 1, 2 or 3 days after isolation of the tissues or organs of origin. In certain embodiments, said cells are cryopreserved after being cultured in a first medium as described in section 6.2.1 above, by approximately 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 or 28 days. In some embodiments, said cells are cryopreserved after being cultured in a first medium as described in section 6.2.1 above, by approximately 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 25, 27 or 28, in a second medium by approximately 1, 2, 3, 4, 5, 6, 7, 8, 9, 0, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 or 28 days as described in section 6.2.1 above.
In one aspect, a method of cryopreservating a population of NK cells, e.g., TSNK cells, is provided herein. In one embodiment, said method comprises: (a) sowing a population of hematopoietic stem or progenitor cells in a first medium comprising interleukin-1 L (IL-15) and, optionally, one or more of the stem cell factor (SCF) and interleukin-7 (IL-7), wherein said optional IL-15 and SCF and IL-7 are not comprised within an undefined component of said medium, so that the population expands, and a plurality of stem cells or hematopoietic progenitors within said population of hematopoietic stem or progenitor cells differentiate into NK cells during said expansion; (b) expanding the cells of step (a) in a second medium comprising interleukin-2 (IL-2), to produce a population of activated NK cells, and (c) cryopreservating the NK cells of step (b) in a means of cryopreservation. In a specific embodiment, said step (c) further comprises (1) preparing a cell suspension solution; (2) adding cryopreservation medium to the cell suspension solution of step (1) to obtain cryopreserved cell suspension; (3) cooling the cryopreserved cell suspension of step (3) to obtain a cryopreserved sample; and (4) store the cryopreserved sample below -80 ° C. In certain embodiments, the method includes any intermediate step between step (a) and (b), and between step (b) and (c), and / or no additional culture step before step (a).
In another embodiment, said method of cryopreservating a population of NK cells, eg, TSNK cells comprises: (a) expanding a population of hematopoietic stem or progenitor cells into a first medium comprising one or more of the stem cell factor (SCF) , IL-2, interleukin-7 (IL-7), interleukin-15 (IL-15) and heparin, and wherein said SCF, IL-2, IL-7 and IL-15 are not comprised within a non-component. defined from said medium, and wherein a plurality of hematopoietic stem or progenitor cells within said population of hematopoietic stem or progenitor cells differentiate into NK cells during said expansion; (b) expanding the cells of step (a) in a second medium comprising interleukin-2 (IL-2), to produce a population of activated NK cells, and (c) cryopreservating the NK cells of step (b) in a means of cryopreservation. In a specific embodiment, said step (c) further comprises (1) preparing a cell suspension solution; (2) adding cryopreservation medium to the cell suspension solution of step (1) to obtain cryopreserved cell suspension; (3) cooling the cryopreserved cell suspension of step (3) to obtain a cryopreserved sample; and (4) classify the cryopreserved sample below -80 ° C. In certain embodiments, the method includes any intermediate step between step (a) and (b), and between step (b) and (c).
The cells provided herein are preferably cooled in a controlled rate freezer, for example, to about 0.1, 0.3, 0.5 or 1 ° C / min during the cryopreservation A preferred cryopreservation temperature is from about -80 ° C to about -180 ° C, preferably from about -125 ° C to about -140 ° C. The cryopreserved cells can be transferred to liquid nitrogen before thawing to be used. In some embodiments, for example, once the ampoules reach approximately -90 ° C, they are transferred to a liquid nitrogen storage area. The cryopreserved cells are preferably thawed at a temperature of about 25 ° C to about 40 ° C, preferably at a temperature of about 37 ° C. In certain embodiments, the cryopreserved cells are thawed after being cryopreserved for approximately 1, 2, 4, 6, 10, 12, 18, 20 or 24 hours, or for approximately 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 or 28 days. In certain embodiments, the cryopreserved cells are thawed after being cryopreserved for approximately 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 or 28 months. In certain embodiments, the cryopreserved cells are thawed after being cryopreserved for approximately 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 years.
Suitable de-icing medium includes, but is not limited to, normal saline, plasma culture medium including, for example, growth medium, eg, RPMI medium. In preferred embodiments, the thawing medium comprises one or more medium supplements (e.g., nutrients, cytokines and / or factors). Supplements medium suitable for thawing cells provided herein include, for example, without limitation, serum such as human AB serum, fetal bovine serum (FBS) or fetal calf serum (FCS), vitamins, human serum albumin (HSA ), bovine serum albumin (BSA), amino acids (e.g., L-glutamine), fatty acids (e.g., oleic acid, linoleic acid or palmitic acid), insulin (e.g., recombinant human insulin), transferrin (human transferrin iron saturated), beta-mercaptoethanol, stem cell factor (SCF), ligand tyrosine kinase 3 Fms (FU3-L), cytokines such as interleukin-2 (IL - 2), interleukin-7 (IL-7 ), interleukin-15 (IL-15), thrombopoietin (Tpo) or heparin. In a specific embodiment, the thawing medium useful in the methods provided herein comprises RPMI. In another specific embodiment, said defrosting means comprises plasmalite. In another specific embodiment, said defrosting means comprises approximately 0.5-20% FBS. In another specific embodiment, said defrosting means comprises about 1, 2, 5, 10, 15 or 20% FBS. In another specific embodiment, said defrosting means comprises approximately 0.5-20% HSA. In another specific embodiment, said defrosting means comprises about 1, 2.5, 5, 10, 15 or 20% HSA. In a more specific embodiment, said defrosting means comprises RPMI and approximately 10% FBS. In another more specific embodiment, said means of thawing comprises plasmalite and about 5% HSA.
The methods of cryopreservation provided herein can be optimized to allow long-term storage, or under conditions that inhibit cell death, for example, by apoptosis and necrosis. In one embodiment, the post-thawing cells comprise more than 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 98% of viable cells, as determined, for example, by Automatic cellular counter or tryptan blue method. In another embodiment, post-thawing cells comprise about 0.5, 1, 5, 10, 15, 20 or 25% of dead cells. In another embodiment, post-thawing cells comprise about 0.5, 1, 5, 10, 15, 20 or 25% of early apoptotic cells. In another modality, approximately 0.5, 1, 5, 10, 15 or 20% of post-thawing cells undergo apoptosis after 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 , 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 or 28 days after thawing, for example, as determined by an apoptosis test (for example, TO-PR03 or AnnV / PI Apoptosis assay kit). In certain embodiments, the post-thawing cells are re-cryopreserved after being cultured, expanded or differentiated using methods provided herein. 6. 8. Uses of TSNK Cells The TSNK cells provided herein are used in methods of treating individuals having cancer, for example, individuals having solid tumor cells and / or blood cells with cancer, or persons having a viral infection. The TSNK cells provided herein can also be used in methods of suppressing the proliferation of tumor cells. 6. 8.1. Treatment of Individuals with Cancer In one embodiment, a method of treating an individual having a cancer, for example, a blood cancer or a solid tumor, which comprises administering to said individual a therapeutically effective amount of TSNK cells is provided. In certain embodiments, the individual has a deficiency of natural killer cells, for example, a deficiency of NK cells active against the individual's cancer. In a specific modality, the method further comprises administering to said isolated perfusate of isolated placenta or isolated placental perfusate cells, for example, a therapeutically effective amount of placental perfusate or isolated placental perfusate cells. In another specific embodiment, the method further comprises administering to said subject an effective amount of an immunomodulatory compound, for example, an immunomodulatory compound. described in section 6.2.1, earlier, or thalidomide. As used herein, an "effective amount" is an amount that, for example, results in a detectable improvement of, reducing the progress of, or eliminating, one or more symptoms of a cancer from which the individual suffers.
In a specific embodiment, the cancer is blood cancer, for example, a leukemia or a lymphoma. In more specific modalities, the cancer is an acute leukemia, for example, acute T cell leukemia, acute myelogenous leukemia (AL), acute promyelocytic leukemia, acute myeloblastic leukemia, acute megakaryoblastic leukemia, B-precursor acute lymphoblastic leukemia, acute lymphoblastic leukemia of precursor T, Burkitt's leukemia (Burkitt's lymphoma), or acute biphenotypic leukemia; a chronic leukemia, for example, chronic myeloid lymphoma, chronic myelogenous leukemia (CML), chronic monocytic leukemia, chronic lymphocytic leukemia (CLL) / small lymphocytic lymphoma, or prolificcytic B-cell leukemia; hairy cell lymphoma; T-cell prolymphocytic leukemia; or a lymphoma (e.g., histiocytic lymphoma, lymphoplasmacytic lymphoma (e.g., Waldenstrom's macroglobulinemia), splenic marginal zone lymphoma, plasma cell neoplasm (e.g., plasma cell myeloma, plasmacytoma, an immunoglobulin deposition disease) monoclonal, or a heavy chain disease), extranodal marginal zone B-cell lymphoma (MALT lymphoma), nodal marginal zone B-cell lymphoma (NMZL), follicular lymphoma, mantle cell lymphoma, diffuse large B-cell lymphoma , large mediatinal (thymic) B-cell lymphoma, large intravascular B-cell lymphoma, primary effusion lymphoma, large granular T-cell lymphocytic leukemia, aggressive NK cell leukemia, adult T-cell leukemia / lymphoma, NK cell lymphoma / Extranodal T, nasal type, enteropathy type T cell lymphoma, hepatosplenic T-cell lymphoma, blastic NK cell lymphoma, mycosis fungoides (Sezary syndrome) ), a primary cutaneous CD30-positive T cell lymphoproliferative disorder (eg, primary cutaneous anaplastic large cell lymphoma or lymphomatoid papulosis), angioimmunoblastic T cell lymphoma, peripheral T cell lymphoma, unspecified, anaplastic large cell lymphoma, a Hodgkin's lymphoma or a predominant nodular lymphocyte Hodgkin's lymphoma. In another specific modality, the cancer is multiple myeloma or myelodysplastic syndrome.
In certain other specific embodiments, the cancer is a solid tumor, for example, a carcinoma, such as an adenocarcinoma, an adrenocortical carcinoma, a colon adenocarcinoma, a colorectal adenocarcinoma, a colorectal carcinoma, a ductal cell carcinoma, a carcinoma of the lung, a thyroid carcinoma, a nasopharynx carcinoma, a melanoma (for example, a malignant melanoma), a nonmelanoma skin carcinoma, or an unspecified carcinoma; a tumor unmolds; a small demoplastic round cell tumor; an endocrine tumor; an Ewing's sarcoma; a germ cell tumor (eg, testicular cancer, ovarian cancer, choriocarcinoma, endodermal sinus tumor, germinoma, etc.); a hepatosblastoma; a hepatocellular carcinoma; a neuroblastoma; a soft tissue sarcoma of no rhabdomyosarcoma; an osteosarcoma; a retinoblastoma; a rhabdomyosarcoma; or a Wilms tumor. In another embodiment, the solid tumor is pancreatic cancer or breast cancer. In other modalities, the solid tumor is an acoustic neuroma; an astrocytoma (for example, a grade I pilocytic astrocytoma, a grade II low grade astrocytoma, a grade III anaplastic astrocytoma, or a grade IV glioblastoma multiforme); a chordoma; a craniopharyngioma; a glioma (for example, a glioma of the brainstem, an ependymoma, a mixed glioma, an optic nerve glioma, or a subependymoma); a glioblastoma; a medulloblastoma; a meningioma; a metastatic brain tumor; an oligodendroglioma; a pineoblastoma; a pituitary tumor; a primitive neurorectodermal tumor; or a schwannoma. In another modality, cancer is prostate cancer.
In certain embodiments, the individual having a cancer, for example, a blood cancer or a solid tumor, for example, an individual who has a deficiency of natural killer cells, is an individual who has received a bone marrow transplant before said bone marrow transplant. administration. In certain modalities, the bone marrow transplant was in treatment of said cancer. In certain other modalities, the bone marrow transplant was under treatment for a condition other than said cancer. In certain embodiments, the individual received an immunosuppressant in addition to said bone marrow transplant. In certain embodiments, the individual who has had a bone marrow transplant exhibits one p plus symptoms of graft-versus-host disease (GVHD) at the time of said administration. In certain other embodiments, the individual who has had a bone marrow transplant is administered said cells before a symptom of graft-versus-host disease (GVHD) is manifested.
In certain specific embodiments, the individual having a cancer, e.g., a blood cancer, has received at least one dose of a TNFa inhibitor, e.g., ETANERCEPT® (Enbrel), prior to said administration. In specific embodiments, said individual received said dose of a TNFa inhibitor within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 months of diagnosis of said cancer. In a specific embodiment, the individual who has received a dose of a TNFa inhibitor exhibits acute myeloid leukemia. In a more specific embodiment, the individual who has received a dose of a TNFa inhibitor and exhibits acute myeloid leukemia also exhibits omission of the long arm of chromosome 5 in globules. In another embodiment, the individual having a cancer, for example, a blood cancer, exhibits a Philadelphia chromosome.
In certain other embodiments, the cancer, for example, a blood cancer or a solid tumor, in said individual is refractory to one or more anti-cancer drugs. In a specific modality, the cancer is refractory to GLEEVEC® (imatinib mesylate).
In certain embodiments, the cancer, for example, a blood cancer, in said individual responds to at least one anti-cancer drug; in this embodiment, perfused placenta, isolated placental perfusate cells, isolated natural killer cells, e.g., natural placental killer cells, e.g., natural killer cells derived from placental, isolated combined natural killer cells, and / or combinations thereof, and optionally an immunomodulatory compound, are added as adjunctive treatments or as a combination therapy with said anti-cancer drug. In certain other embodiments, the individual having a cancer, for example, a blood cancer, has been treated with at least one anti-cancer drug, and has relapsed, prior to said administration.
In one aspect, herein is provided a method of treating an individual having multiple myeloma, comprising administering to the individual (1) lenalidomide; (2) melphalan; and (3) expanded NK cells, wherein said NK cells are effective to treat multiple myeloma in said individual. In a specific embodiment, said NK cells are NK cells of cord blood, or NK cells produced from cord blood hematopoietic cells, e.g., hematopoietic stem cells. In another embodiment, said NK cells have been produced by any of the methods described herein to produce NK cells, for example, to produce TSNK cells. In another embodiment, said NK cells have been expanded prior to said administration. In another embodiment, said lenalidomide, melphalan and / or NK cells are administered separately from each other. In certain specific embodiments of the method of treating an individual with multiple myeloma, said NK cells are produced by a method comprising: expanding a population of hematopoietic stem or progenitor cells into a first medium comprising one or more of the stem cell factor (SCF) ), IL-2, interleukin-7 (IL-7), interleukin-15 (IL-15) and heparin, and wherein said SCF, IL-2, IL-7 and IL-15 are not included within a component undefined of said medium, and wherein a plurality of stem cells or hematopoietic progenitors within said population of stem cells or hematopoietic progenitors differentiate into NK cells during said expansion; and expanding the cells of step (a) in a second medium comprising interleukin-2 (IL-2).
In another aspect, a method of treating an individual having chronic lymphocytic leukemia (CLL), which comprises administering to the individual a therapeutically effective dose of (1) lenalidomide, is provided; (2) melphalan; (3) Fludarabine; and (4) expanded NK cells, for example, TSNK cells, wherein said NK cells are cord blood NK cells, or NK cells produced from cord blood hematopoietic stem cells. In another embodiment, said NK cells have been produced by any of the methods described herein to produce NK cells, for example, to produce TSNK cells. In a specific embodiment of any of the above methods, said NK cells have been expanded for at least 10 days before said administration. In a specific embodiment of any of the above methods, said lenalidomide, melphalan, fludarabine and expanded NK cells are administered to said individual separately. In certain specific embodiments of the method of treating an individual with CLL, said NK cells are produced by a method comprising: expanding a population of stem cells or hematopoietic progenitors in a first medium comprising one or more of the stem cell factor (SCF) , IL-2, interleukin-7 (IL-7), interleukin-15 (IL-15) and heparin, and wherein said SCF, IL-2, IL-7 and IL-15 are not comprised within a non-component. defined from said medium, and wherein a plurality of stem cells or hematopoietic progenitors within said population of stem cells or hematopoietic progenitors differentiate into NK cells during said expansion; and expanding the cells of step (a) in a second medium comprising interleukin-2 (IL-2), to produce activated NK cells. 6. 8.2. Suppression of Tumor Cell Proliferation In addition, a method of suppressing the proliferation of tumor cells, which comprises contacting the tumor cells with TSNK cells, is provided herein. Optionally, the tumor cells and / or TSNK cells are contacted with isolated placental perfusate or isolated placental perfusate cells. In another specific embodiment, the tumor cells and / or TSNK cells are additionally contacted with an immunomodulatory compound, for example, an immunomodulatory compound described in section 6.2.1, above, or thalidomide, so that the proliferation of the tumor cells is reduces detectably compared to tumor cells of the same type not contacted with TSNK cells. Optionally, the tumor cells and / or TSNK cells contacted with an immunomodulatory compound are contacted with isolated placental perfusate or isolated placental perfusate cells.
As used herein, "contact" with respect to cells, in a modality encompasses direct physical contact, eg, cell-cell, between placental perfusate, placental perfusate cells, natural killer cells, eg, TSNK cells , and / or isolated combined natural killer cells and tumor cells. In another embodiment, "contact" encompasses presence in the same physical space, for example, perfused placenta, placental perfusate cells, natural killer cells, for example, natural killer cells intermediate of placenta, and / or combined natural killer cells isolated they are placed in the same container, for example, culture dish, multi-well plate) as tumor cells. In another embodiment, "contacting" perfusate of placenta, placental perfusate cells, combined natural killer cells or natural killer cells intermediate of placenta, and tumor cells is achieved, for example, by injecting or infusing the perfusate of placenta or cells, by example, placental perfusate cells, combined natural killer cells or natural killer cells, eg, natural killer cells intermediate placenta in an individual, eg, a human comprising tumor cells, eg, a cancer patient. "Contact" in the context of immunomodulatory compounds and / or thalidomide are physically contacted with each other, or are placed within the same physical volume (e.g., a cell culture container or an individual).
In a specific embodiment, the tumor cells are blood cancer cells, for example, leukemia cells or lymphoma cells. In more specific embodiments, the cancer is an acute leukemia, for example, acute T cell leukemia cells, acute myelogenous leukemia (AML) cells, acute promyelocytic leukemia cells, acute myeloblastic leukemia cells, acute megakaryoblastic leukemia cells B-precursor acute lymphoblastic leukemia cells, T-precursor acute lymphoblastic leukemia cells, Burkitt's leukemia cells (Burkitt's lymphoma), or acute biphenotypic leukemia cells; chronic leukemia cells, for example, chronic myeloid lymphoma cells, chronic myelogenous leukemia (CML) cells, chronic monocytic leukemia cells, chronic lymphocytic / small lymphocytic leukemia cells, or B-cell prolymphocytic leukemia cells; hairy cell lymphoma cells; T-cell prolymphocytic leukemia cells; or lymphoma cells, eg, histiocytic lymphoma cells, lymphoplasmacytic lymphoma cells (e.g., Waldenstrom macroglobulinemia cells), splenic marginal zone lymphoma cells, plasma cell neoplasm cells (e.g., myeloma cells) of plasma cell, plasmacytoma cells, monoclonal immunoglobulin deposition disease, or heavy chain disease), extranodal marginal B-cell lymphoma cells (MALT lymphoma), nodal marginal zone B-cell lymphoma cells (NMZL) , follicular lymphoma cells, mantle cell lymphoma cells, large intravascular B cell lymphoma cells, primary effusion lymphoma cells, large T-cell granular lymphocytic leukemia cells, aggressive NK cell leukemia cells, leukemia / adult T cell lymphoma, nasal-type cells - extranodal NK / T cell lymphoma, T-cell lymphoma cells enteropathy, hepatosplenic T cell lymphoma cells, blastic NK cell lymphoma cells, mycosis fungoides (Sezary syndrome), CD30-positive primary cutaneous T cell lymphoproliferative disorder cells (e.g., anaplastic large cell lymphoma) primary cutaneous or lymphomatoid papulosis), angioimmunoblastic T cell lymphoma cells, unspecified peripheral T cell lymphoma cells, nodular lymphocyte predominant Hodgkin lymphoma cells, or Hodgkin lymphoma cells. In another specific embodiment, the tumor cells are multiple myeloma cells or myelodysplastic syndrome cells.
In specific embodiments, the tumor cells are solid tumor cells, e.g., carcinoma cells, e.g., adenocarcinoma cells, adenocortical carcinoma cells, colon adenocarcinoma cells, colorectal adenocarcinoma cells, colorectal carcinoma cells, carcinoma cells. of ductal cell, lung carcinoma cells, thyroid carcinoma cells, nasopharyngeal carcinoma cells, melanoma cells (e.g., malignant melanoma cells), non-melanoma skin carcinoma cells, or unspecified carcinoma cells; tumor cells strips; small desmoplastic round cell tumor cells; endocrine tumor cells; Ewing's sarcoma cells; germ cell tumor cells (e.g., testicular cancer cells, ovarian cancer cells, choriocarcinoma cells, endodermal sinus tumor cells, germinoma cells, etc.); hepatosblastoma cells; hepatocellular carcinoma cells; neuroblastoma cells; soft tissue sarcoma cells from non-rhabdomyosarcoma; osteosarcoma cells; retinoblastoma cells; rhabdomyosarcoma cells; or Wilms tumor cells. In another embodiment, the tumor cells are pancreatic cancer cells or breast cancer cells. In other embodiments, the solid tumor cells are acoustic neuroma cells; astrocytoma cells (eg, grade I polycytic astrocytoma cells, grade II low grade astrocytoma cells, grade III anaplastic astrocytoma cells, or multiform glioblastoma grade IV); chordoma cells; craniofaringioma cells; glioma cells (e.g., brainstem glioma cells; ependymoma cells; mixed glioma cells; optic nerve glioma cells; or subependymoma cells); glioblastoma cells; medulloblastoma cells; meningioma cells; tumor cells of metastatic brain; oligodendroglioma cells; pineoblastoma cells; pituitary tumor cells; primitive neuroectodermal tumor cells; or schwannoma cells. In another embodiment, the tumor cells are prostate cancer cells.
As used herein, "therapeutically beneficial" and "therapeutic benefits" include, but are not limited to, for example, reduction in the size of a tumor; reducing or stopping the expansion of a tumor; reduction in the number of cancer cells in a tissue sample, eg, blood sample, per unit volume; the clinical improvement in any symptom of the particular cancer or tumor said individual has, the reduction or cessation of worsening of any particular cancer symptom the individual has, etc. 6. 8.3. Treatment of cancers using TSNK cells and other anti-cancer agents The treatment of an individual having cancer using the TSNK cells described herein may be part of an anti-cancer therapy regimen that includes one or more other anti-cancer agents. Such anti-cancer agents are well known in the art. Specific anti-cancer agents that can be administered to an individual having cancer, for example, an individual having tumor cells, in addition to the TSNK cells, and optionally perfused, perfusate cells, natural killer cells other than TSNK cells, include, but they are not limited to: acivicin; aclarubicin; benzoyl hydrochloride; Acronine; adozelesina; adrucil; aldesleukin; altretamine; ambomycin; ametantrone acetate; amsacrine; anastrozole; anthramycin; asparaginase; asperlina; avastin (bevacizumab); azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; Sodium brequinar; biririmine; busulfan; cactinomycin; calusterona; caracemide; carbetimer; carboplatin; carmustine; carubicin hydrochloride; carzelesin; cedefingol; celecoxib (COX-2 inhibitor); chlorambucil; Corylemycin; cisplatin; cladribine; crisnatol mesylate; cyclophosphamide; cytarabine; Dacarbazine; Dactinomycin; daunorubicin hydrochloride; decitabine; dexormaplatin; dezaguanine; dezaguanine mesylate; diaziquone; docetaxel; doxorubicin; Doxorubicin hydrochloride; droloxifene; Droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflomitin hydrochloride; elsamitrucin; enloplatin; enpromato; epipropidine; epirubicin hydrochloride; erbulozole; esorubicin hydrochloride; estramustine; estramustine sodium phosphate; etanidazole; etoposide; etoposide phosphate; etoprin; fadrozole hydrochloride; fazarabine; fenretinide; floxuridine; Fludarabine phosphate; fluorouracil; flurocitabine; fosquidone; fostriecin sodium; gemcitabine; gemcitabine hydrochloride; hydroxyurea; idarubicin hydrochloride; ifosfamide; ilmofosin; iproplatin; Irinotecan; Irinotecan hydrochloride; lanreotide acetate; letrozole; leuprolide acetate; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansina; mechlorethamine hydrochloride; Megestrol acetate; melengestrol acetate; melphalan; menogaril; mercaptopurine; methotrexate; sodium methotrexate; metoprine; meturedepa; mitinomide; mitocarcin; mitochromin; mitogilin; mitomalcin; mitomycin; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nocodazole; nogalamycin; ormaplatin; oxisuran; paclitaxel; pegaspargasa; Peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; plicamycin; pentamethane; sodium porfimer; porphyromycin; Prednimustine; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazof urina; riboprine; safingol; safingol hydrochloride; semustine; simtrazeno; sodium sparfosate; sparsoicin; Spirogermanium hydrochloride; spiromustine; spiroplatin; Streptonigrin; streptozocin; sulofenur; talisomycin; tecogalan sodium; taxotere; tegafur; teloxantrone hydrochloride; temoporfin; teniposide; Teroxirone; testolactone; tiamiprine; thioguanine; thiotepa; thiazofurine; tirapazamine; Toremifene citrate; trestolone acetate; trie i ri b i na phosphate; trimetrexate; trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; Uracil mustard; uredepa; vapreotide; verteporfin; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidin sulfate; sulfate of ving licinato; vinleurosine sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorozole; zipiplatine; zinostatin; and zorubicin hydrochloride.
Other anti-cancer drugs include, but are not limited to: 20-epi-1,25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acilfulveno; adecipenol; adozelesina; adlesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrografol; inhibitors of angiogenesis; antagonist D; antagonist G; antarelix; anti-dorsalizing morphogenetic protein-1; antiandrogen; Prostatic carcinoma; antiestrogen; antineoplaston; anti-sense oligonucleotides; afidicolin glycinate; modulators of apoptosis genes; apoptosis regulators; Apurinic acid; ara-CDP-DL-PTBA; arginine deaminase; asulacrine; atamestane; atrimustine; axinastatin 1; axinastatin 2; axinastatin 3; azasetron; azatoxin; azathirosine; Baccatin III derivatives; balanol batimastat; BCR / ABL antagonists; benzoclorins; benzoylstaurosporin; beta lactam derivatives; beta-aletine; betaclamycin B; betulinic acid; bFGF inhibitor; bicalutamide; bisantrene; bisaziridinyl espermine; bisnafida; bistratene A; bizelesin; breflato; biririmine; budotitan; butionine sulfoximine; calcipotriol; calfostin C; camptosar (also called Campto; irinotecan) derived from camptothecin; capecitabine; carboxamide-amino-triazole; carboxyamidotriazole; CaRest M3; CARN 700; inhibitor derived from cartilage; carzelesin; casein kinase inhibitors (ICOS); castanospermine; cecropin B; cetrorelix; chlorines; Chloroquinoxaline sulfonamide; cycaprost; cis-porphyrin; cladribine; clomiphene analogues; clotrimazole; crambescidin 816; crisnatol; cryptophycin 8; Cryptophycin A derivatives; curacin A; cyclopentantraquinones; Cycloplatam; cipemycin; cytarabine ocphosphate; cytolytic factor; cytostatin; dacliximab; decitabine; dehydrodidenmine B; deslorelin; dexamethasone; dexiphosphamide; dexrazoxane; dexverapamil; diaziquone; didemnin B; didox; diethylnorspermine; dihydro-5-azacytidine; dihydrotaxol, 9-; dioxamycin; diphenyl spiromustine; docetaxel; docosanol; dolasetron; doxifluridine; doxorubicin; droloxifene; dronabinol; duocarmicin SA; ebselen; ecomustine; edelfosin; Edrecolomab; eflornithine; elemeno; emitefur; epirubicin; epristerida; estramustine analogue; estrogen agonists; estrogen antagonists; etanidazole; etoposide phosphate; exemestane; fadrozole; fazarabine; fenretinide; filgrastim; Finasteride; vopiridol; flezelastine; fluasterone; fludarabine; fluorodaunorunicin hydrochloride; forfenimex; formestane; fostriecin; fotemustine; gadolinium texaphyrin; gallium nitrate; galocitabine; ganirelix; gelatinase inhibitors; gemcitabine; glutathione inhibitors; hepsulfam; heregulina; hexamethylene bisacetamide; hypericin; ibandronic acid; idarubicin; idoxifen; idramantone; ilmofosin; ilomastat; imatinib (for example, GELLEVE®), imiquimod; immunostimulatory peptides; inhibitor of growth factor 1 receptor as insulin; interferon agonists; interferons; interleukins; iobenguan; iododoxorubicin; ipomeanol, 4-; iroplact; irsogladine; Sobengazol; isohomohalicondrine B; itasetron; jasplakinolide; kahalalide F; lamelarin-N triacetate; lanreotide; leinamycin; lenograstim; lentinan sulfate; leptolstatin; letrozole; Leukemia inhibitory factor; leukocyte interferon alpha; leuprolide + estrogen + progesterone; leuprorelin; levamisole; liarozole; linear polyamine analog; lipophilic disaccharide peptide; lipophilic platinum compounds; lisoclinamide-7; lobaplatin; lombrlcina; lometrexol; lonidamine; losoxantrone; loxoribine; lurtotecan; lutetium texaphyrin; lyophilin; UTIC peptides; Maytansine; Handstatin A; marimastat, masoprocol; maspina; matrilysin inhibitors; matrix metalloproteinase inhibitors; menogaril; merbarone; meterelin; methioninase; metoclopramide; inhibitor MY F; mifepristone; miltefosine; mirimostim; mitoguazone; mitolactol; mitomycin analogues; mitonafide; saporin growth factor of mitotoxin filbroblast; mitoxantrone; mofarotene; molgramostim; Erbitux (cetuximab), human chorionic gonadotropin; lipid A of monophosphoryl + myobacterial cell wall sk; mopidamol; mustard anti-cancer agent; micaperoxide B; mycobacterial cell wall extract; myriaporone; N-acetyldinaline; N-substituted benzamides; nafarelin; nagrestip; naloxone + pentazocine; napavina; nafterpina; nartograstim; nedaplatin; nemorubicin; acid neridronic; nilutamide; nisamycin; Nitric oxide modulators; nitroxide antioxidant; nitrulin; oblimersen (GENASENSE®); O6-benzylguanine; octreotide; okicenona; oligonucleotides; onapristone; ondansetron; oracine; oral cytosine inducer; ormaplatin; osaterone; oxaliplatin (e.g., Floxatin); oxaunomycin; paclitaxel; Paclitaxel analogues; paclitaxel derivatives; palauamine; palmitoylrhizoxin; pamidronic acid; panaxitriol; panomiphene; parabactin pazeliptina; pegaspargasa; peldesina; pentosan sodium polulose; pentostatin; pentrozole; perflubron; perfosfamide; perilyl alcohol; phenazinomycin; phenylacetate; phosphatase inhibitors; picibanil; pilocarpine hydrochloride; pirarubicin; piritrexim; placetina A; placetina B; inhibitor plasminogen activator; platinum complex; platinum compounds; platinum-triamine complex; sodium porfimer; porphyromycin; prednisone; propyl bis-acridone; prostaglandin J2; proteasome inhibitors; immune modulator based on protein A; inhibitor of protein kinase C; inhibitors of protein kinase C; microalgal; protein tyrosine phosphatase inhibitors; inhibitors of purine nucleoside phosphorylase; purpurins; pyrazoloacridine; polyoxyethylene conjugate of pyridoxylated hemoglobin; raf antagonists; raltitrexed; ramosetron; ras farnesyl ras transferase protein inhibitors; ras inhibitors; ras-GAP inhibitor; Demethylated reteliptine; Re 186 rhenium etidronate; rhizoxin; ribozymes; retinamide Rll; rohituquine; romurtida; roquinimex; Rubiginone B1; ruboxyl; safingol; saintopine; SarCNU; sarcofitol A; sagramostim; Sdi 1 mimetic; semustine; inhibitor derived from senescence 1; sense oligonucleotides; inhibitors of signal transduction; sizofiran; Sobuzoxane; sodium borocaptate; sodium phenylacetate; solverol; Somatomedin binding protein; sonermin; Sparfosic acid; spicamycin D; spiromustine; splenopentin; spongistatin 1; squalamine; stipiamide; stromelsin inhibitors; Sulfinosine; superactive vasoactive intestinal peptide antagonist; suradista suramin; Swainsonin; talimustine; tamoxifen methylodide; tauromustine; tazarotene; tecogalan sodium; tegafur; telurapyrilio; telomerase inhibitors; temoporfin; teniposide; tetrachlorodeoxide, tetrazomine; Taliblastine; thiocoraline; thrombopoietin; mimetic thrombopoietin; timalfasin; thymopoietin receptor agonist; thymotrinam; thyroid stimulating hormone; tin ethyl etpurpurine; tirapazamine; titanocene bichloride; topsentin; toremifene; Translation inhibitors; tretinoin; triacetyluridine; triciribine; trimetrexate; triptorelin; tropisetron; turosteride; tyrosine kinase inhibitors; Tyrphostins; UBC inhibitors; ubenimex; growth inhibitory factor derived from urogenital sinus; Urokinase receptor antagonists; vapreotide; Variolin B; Vectibix (panitumumab) velaresol; veramina; verdinas; verteporfin; vinorelbine; vinxaltine; vitaxin; vorozole; Welcovorin (leucovorin); Xeloda (capecitabine); zanoterone; zipiplatine; zilascorb; and zinostatin estimalmer. 6. 8.4. Viral Infection Treatment In another embodiment, a method of treating an individual having a viral infection, which comprises administering to said individual a therapeutically effective amount of TSNK cells, is provided herein. In certain embodiments, the individual has a deficiency of natural killer cells, for example, a deficiency of active NK cells against the individual's viral infection. In certain specific embodiments, said administration further comprises administering to the individual one or more of placental perfusate isolated, isolated placental perfusate cells, isolated natural killer cells, eg, natural killer cells of placenta, eg, derived intermediate natural killer cells of placenta, isolated combined natural killer cells and / or combinations thereof. In certain specific modalities, TSNK cells are contacted with an immunomodulatory compound, for example, an immunomodulatory compound described in 6.2.1, above, or thalidomide, prior to said administration. In certain other specific embodiments, said administration comprises administering an immunomodulatory compound, for example, an immunomodulatory compound described in section 6.2.1, above, or thalidomide, to said individual in addition to said TSNK cells, wherein said amount is an amount that is , for example, results in a detectable improvement of, reducing the progress of, or elimination of, one or more symptoms of said viral infection. In specific modalities, the viral infection is an infection by a virus of the family Adenoviridae, Picornaviridae, Herpesviridae, Hepadnaviridae, Flaviviridae, Retroviridae, Orthomyxoviridae, Paramyxoviridae, Papilommaviridae, Rhabdoviridae or Togaviridae. In more specific modalities, said virus is human immunodeficiency virus (HIV), coxsackievirus, hepatitis A virus (HAV), poliovirus, Esptein-Barr virus (EBV), herpes simplex type 1 (HSV1), herpes simplex type 2 (HSV2), human cytomegalovirus (CMV), human herpesvirus type 8 (HHV8), herpes zoster virus (varicella zoster virus (VZV) or herpes virus), hepatitis B virus (HBV), hepatitis C virus (HCV) , hepatitis D virus (HDV), hepatitis E virus (HEV), influenza virus (for example, influenza A virus, influenza B virus, influenza C virus or togotovirus), measles virus, mumps virus, parainfluenza virus, papilloma virus, rabies virus or rubella virus.
In other more specific embodiments, said virus is adenovirus species A, serotype 12, 18 or 31; adenovirus species B, serotype 3, 7, 11, 14, 16, 34, 35 or 50; adenovirus species C, serotype 1, 2, 5 or 6; species D, serotype 8, 9, 10, 13, 15, 17, 19, 20, 22, 23, 24, 25, 26, 27, 28, 29, 30, 32, 33, 36, 37, 38, 39, 42, 43, 44, 45, 46, 47, 48, 49 or 51; species E, serotype 4; or species F, serotype 40 or 41.
In certain other more specific modalities, the virus is Apoi virus (APOIV), Aroa virus (AROAV), bagasse virus (BAGV), Banzi virus (BANV), Bouboui virus (BOUV), Cacipacore virus (CPCV), Carey Island virus ( CIV), Cowbone Ridge virus (CRV), Dengue virus (DENV), Edge Hill virus (EHV), Gully Gadget virus (GGYV), llheus virus (ILHV), Israel turkey meningoencephalomyelitis virus (ITV), encephalitis virus Japanese (JEV), Jugra virus (JUGV), Jutiapa virus (JUTV), Kadam virus (KADV), Kedougou virus (KEDV), Kokobera virus (KOKV), Koutango virus (KOUV), Kyasanur Forest disease virus (KFDV), Langat virus (LGTV), Meaban virus (MEAV), Modoc virus (MODV), Montana myotis leukoencephalitis virus (MMLV), Murray Valley encephalitis virus (MVEV), Ntaya virus (NTAV), Omsk hemorrhagic fever virus (OHFV) ), Powassan virus (POWV), Rio Bravo virus (RBV), Royal Pharmaceutically acceptable virus (RFV), Savoy virus (SABV), St. Louis encephalitis virus (SLEV), Sal Veja virus (SVV), San Perlita virus (SPV), Saumarez Reef virus (SREV), Sepik virus (SEPV), Tembusu virus (TMUV), tick-borne encephalitis virus (TBEV), Tyuleniy virus (TYUV), Uganda S virus (UGSV), virus Usutu (USUV), Wesselsbron virus (WESSV), West Nile virus (WNV), Yaounde virus (YAOV), yellow fever virus (YFV), Yokose virus (YOKV) or Zika virus (ZIKV).
In other embodiments, TSNK cells, and optionally perfused placenta and / or perfusate cells, are administered to an individual having a viral infection as part of an antiviral therapy regimen that includes one or more other antiviral agents. Specific antiviral agents that can be administered to an individual having a viral infection include, but are not limited to: imimoimod podofilox, podophyllin, interferon alpha (IFNa), lattices, nonoxynol-8, acyclovir, famciclovir, valaciclovir, ganciclovir, cidofovir; amantadine, rimantadine; ribavirin; zanamavir and oseltaumavir; protease inhibitors such as indinavir, nelfinavir, ritonavir or saquinavir; inhibitors of inverted nucleoside transcriptase such as didanosine, lamivudine, stavudine, zalcitabine or zidovudine; and non-nucleoside inverted transcriptase inhibitors such as neviparin or efavirenz. 6. 8.5. Administration The determination of the number of cells, for example, placental perfusate cells, for example, placental perfused nucleated cells, combined natural killer cells, and / or isolated natural killer cells, e.g., TSNK cells, and quantity determination of an immunomodulatory compound, for example, as an immunomodulatory compound in section 6.2.1, above, or thalidomide, can be performed independently of one another. 6. 8.5.1. Cell Administration In certain embodiments, TSNK cells are used, for example, administered to an individual, in any amount or number that results in a detectable therapeutic benefit to the individual, for example, an effective amount, wherein the individual has a viral infection, cancer or tumor cells, for example, an individual having tumor cells, a solid tumor or a blood cancer, for example, a patient with cancer. Said cells can be administered to said individual by absolute numbers of cells, for example, said individual can be administered at about, at least approximately, or at most approximately, 1 x 105, 5 x 105, 1 x 106, 5 x 106 , 1 x 107, 5 x 107, 1 x 108, 5 x 108, 1 X 109, 5 x 109, 1 x 1010, 5 x 1010 or 1 x 1011 TSNK cells. In other embodiments, the TSNK cells can be administered to said individual by relative numbers of cells, for example, said individual can be administered at about, at least approximately, or at most approximately, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108, 1 x 109, 5 x 109, 1 x 1010, 5 x 1010 or 1 x 1011 TSNK cells per kilogram of the individual. TSNK cells can be administered to said individual according to an approximate ratio between a number of TSNK cells, and optionally placental perfusate cells and / or natural killer cells other than TSNK cells, and a number of tumor cells in said individual (eg, example, an estimated number). For example, TSNK cells can be administered to said individual in a ratio of about, at least about or at most about 1: 1, 2: 1, 3: 1, 4: 1, 5: 1, 6: 1, 7: 1, 8: 1, 9: 1, 10: 1, 15: 1, 20: 1, 25: 1, 30: 1, 35: 1, 40: 1, 45: 1, 50: 1, 55: 1, 60: 1, 65: 1, 70: 1, 75: 1, 80: 1, 85: 1, 90: 1, 95: 1 or 100: 1 to the number of tumor cells in the individual. The number of cells Tumor in said individual can be estimated, for example, by counting the number of tumor cells in a tissue sample from the individual, for example, blood sample, biopsy, or the like. In specific modalities, for example, for solid tumors, said counting is performed in combination with images of the tumor or tumors to obtain an approximate tumor volume. In a specific embodiment, an immunomodulatory compound or talimodomide, for example, an effective amount of an immunomodulatory compound or thalidomide, are administered to the individual in addition to the TSNK cells, optionally placental perfusate cells and / or natural killer cells other than the TSNK cells.
In certain embodiments, the method of suppressing the proliferation of tumor cells, for example, in an individual; treatment of an individual having a deficiency in the natural killer cells of the individual; or treatment of an individual having tumor cells, a blood cancer or a solid tumor, comprises contacting the tumor cells, or administering to said individual, a combination of TSNK cells and one or more perfusate of placenta and / or perfusate cells of placenta. In specific embodiments, the method comprises additionally contacting the tumor cells, or administering to the individual, an immunomodulatory compound or thalidomide.
In a specific embodiment, for example, treatment of an individual having a deficiency in the natural killer cells of the individual (eg, a deficiency in the number of NK cells or in the reactivity of NK cells to a cancer, tumor or cell virally infected); or treatment of an individual having a cancer or a viral infection, or suppression of tumor cell proliferation, comprising contacting said tumor cells, or administering to said individual, TSNK cells supplemented with placental perfusate cells isolated or perfused from placenta. In specific modalities, approximately, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108 or more TSNK cells per milliliter, or, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108, 1 x 109, 5 x 109, 1 x 1010, 5 x 1010 or 1 x 1011 or more TSNK cells, are supplemented with approximately, or at least approximately, 1 x 105, 5 x 105, 1 x 106 , 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108 or more isolated placental perfusate cells per milliliter, or, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108, 1 x 109, 5 x 109, 1 x 1010, 5 x 1010 or 1 x 1011 0 more isolated placental perfusate cells. In other more specific modalities, approximately 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108 or more TSNK cells per milliliter, or, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 X 108, 5 x 108, 1 x 109, 5 x 109, 1? 1010, 5? 1010 or 1 x 1011 or more TSNK cells are supplemented with approximately, or at least approximately, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950 or 1000 ml_ of perfusate, or approximately 1 unit of perfusate.
In another specific embodiment, the treatment of an individual having a deficiency in the natural killer cells of the individual; treatment of an individual who has cancer; treatment of an individual having a viral infection; or suppression of tumor cell proliferation, comprises contacting the tumor cells, or administering to the individual, TSNK cells, wherein said cells are supplemented with adherent placental cells, for example, placental perfused stem cells or multipotent cells, for example, Placental cells adherent to plastic culture CD34 \ CD10 \ CD105 +, CD200 +. In specific embodiments, TSNK cells are supplemented with approximately 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 1 O8 or more placental stem cells adherent per milliliter, or, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108, 1 x 109, 5 x 109, 1 x 1010, 5 x 1010 or 1 x 1011 or more adherent placental cells, e.g., adherent placental stem cells or multipotent cells.
In another specific embodiment, the treatment of an individual having a deficiency in the natural killer cells of the individual; treatment of an individual who has cancer; treatment of an individual who has a viral infection; or suppression of tumor cell proliferation, is performed using an immunomodulatory compound or thalidomide in combination with TSNK cells, in wherein said cells are supplemented with conditioned medium, for example, conditioned medium by placental cells adherent to tissue culture plastic CD34", CD10 +, CD105 +, CD200 +, for example, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6 , 0.7, 0.8, 0.9, 1. 2, 3, 4, 5, 6, 7, 8, 9, 10 mL of culture medium conditioned by stem cell per perfusate unit, or by 104, 105, 106, 107, 108, 109, 1010 or 1011 TSNK cells In certain embodiments, the tissue-adherent plastic placenta cells are the multipotent adherent placental cells described in US Patent No. 7,468,276 and US Patent Application Publication No. 2007/0275362, the descriptions of which are incorporated herein by reference in their entireties In another specific embodiment, the method further comprises contacting the tumor cells, or administering to the individual, an immunomodulatory compound or thalidomide.
In another specific embodiment, the treatment of an individual having a deficiency in the natural killer cells of the individual; treatment of an individual who has cancer; treatment of an individual who has a viral infection; or suppression of tumor cell proliferation, wherein said TSNK cells are supplemented with placental perfusate cells, the perfusate cells are contacted with interleukin-2 (I L -2) for a period of time before said contact. In certain modalities, said period of time is approximately, at least, or at least much 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40 , 42, 44, 46 or 48 hours before said contact.
TSNK cells, and optionally perfused or perfusate cells, can be administered once to an individual having a viral infection, an individual having cancer, or an individual having tumor cells, during a course of anti-cancer therapy.; or they can be administered multiple times, for example, once every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 , 20, 21, 22 or 23 hours, or once every 1, 2, 3, 4, 5, 6 or 7 days, or once every 1, 2, 3, 4, 5, 6, 7, 8, 9 , 10, 24, 36 or more weeks during therapy. In embodiments, wherein cells and an immunomodulatory compound or thalidomide are used, the immunomodulatory compound or thalidomide, and cells or perfusate, can be administered to the individual together, for example, in the same formulation; separately, for example, in separate formulations, at about the same time; or they can be administered separately, for example, at different dosing schedules or at different times of the day. Similarly, in embodiments where cells and an antiviral compound or anti-cancer compound are used, the antiviral compound or anti-cancer compound, and cells or perfusate, can be administered to the individual together, for example, in the same formulation; separately, for example, in separate formulations, at about the same time; or they can be administered separately, for example, in different dosing schedules or at different times of the day. TSNK cells, and perfused or Perfusate cells can be administered regardless of whether the TSNK, perfused or perfused cells have been administered to the individual in the past. 7. EXAMPLES 7. 1. Example 1: Recovery of Hematopoetic Stem Cells from Human Placenta Perfusate and Umbilical Cord Blood Typically perfused human placenta (HPP) and umbilical cord blood (UCB) were purified using Ficoll and ammonium chloride to obtain total nucleated cells (TNCs). TNCs were then used in a positive selection procedure to isolate CD34 + cells using anti-CD34 and RoboSep spheres according to the manufacturer's protocol (StemCell Technologies, Inc.). In this experiment, CD34 + cells with more than 90% purity were isolated. Alternatively, the EasySep® Human Progenitor Cell Enrichment Kit (StemCell Technologies, Inc.) was used in a negative selection procedure to bypass cells committed to lineage by using Human Progenitor Cell Enrichment Cocktail with monoclonal antibodies to the following human cell surface antigens: CD2, CD3, CD11b, CD11c, CD14, CD16, CD19, CD24, CD56, CD66b and glycophorin A. Using the negative selection, 90% of CD34 + cells were recovered from the raw material. The cellular composition of the recovered HSCs is summarized in Table 1.
Table 1. Cell composition of hematopoietic stem cells (HSCs) enriched. The standard deviation was calculated for the population medium of 3 donors. 7. 2. Example 2: Cell-free Expansion and Differentiation Hematopoietic Stem Cell Feeder in Natural Killer Cells CD34 + cells were cultured in the following medium formulations for up to 48 days, and aliquots of cells were taken for evaluation of cell count, cell viability, characterization of differentiation and functional evaluation of natural killer cells.
NK medium: GBGM (Growth medium based on Glycostem, Glycostem Cat # CCT-SCB500, transparent cellular technology) supplemented with pen / strep (Cat # 15140, Gibco), 20 ng / mL SCF (Cat # 255-SC, R &D Systems), 10 ng / mL ligand Flt-3 (Cat # 308-FK, R &D Systems), 20 ng / mL TPO (Cat # 288-TP, R &D Systems), 20 ng / mL IL -7 (Cat # 207-IL, R &D Systems), 200 lU / mL IL-2 (Cat # 202-IL, R &D Systems) and 10 ng / mL IL-15 (Cat # 247-IL, R &; D Systems).
NK2 medium: DMEM (Cat # MT-10-013-CV, Fisher): Ham's F12 (Cat # BW12-615F, Fisher) as 1: 2 supplemented with 2 mM L-Glutamine (Cat # 25030, Invitrogen), 1% pen / strep, 20% human serum AB (Cat # 100-512, Gemcell), 5 ng / mL sodium selenite (Cat # S9133, Sigma), 50 μ? Ethanolamine (Cat # E0135, Sigma), 25 μ? β-mercaptoethanol (Cat # 21985, Invitrogen), 20 ng / mL ascorbic acid (Cat # 47863, Sigma), 5 ng / mL IL-3 (Cat # 203-IL, R &D Systems), 20 ng / mL SCF , 10 ng / mL ligand Flt-3, 20 ng / mL IL-7 and 10 ng / mL IL-15.
NK3 medium: X-vivo 20 (Cat # BW04-448Q, Fisher) supplemented with pen / strep, 10% human serum AB (Cat # 100-512, Gemcell) and 500 lU / mL IL-2.
NK2A medium: GBGM supplemented with 10% human serum AB, 1% pen / strep, 20 ng / mL SCF, 10 ng / mL ligand Flt-3, 20 ng / mL TPO, 20 ng / mL IL-7, 200 lU / mL IL-2, 10 ng / mL IL-15 and 1.5 lU / mL heparin (Cat # H3149, Sigma).
NK2B1 medium: DMEM: Ham's F12 as 1: 2 supplemented with 2 mM L-glutamine, 1% pen / strep, 20% human AB serum, 5 ng / mL sodium selenite, 50 μ? Ethanolamine, 25 μ? β-mercaptoethanol, 20 μg / mL ascorbic acid, 5 ng / mL IL-3, 20 ng / mL SCF, 10 ng / mL ligand Flt-3, 20 ng / mL IL-7 and 10 ng / mL IL-15.
NK2B2 medium: DMEM: Ham's F12 as 1: 2 supplemented with 2 (IIM L-glutamine, 1% pen / strep, 20% human AB serum, 5 ng / mL sodium selenite, 50 μ? Ethanolamine, 25 μ? β-mercaptoethanol, 20 μg / mL ascorbic acid, 200 μU / mL IL-2, 20 ng / mL SCF, 10 ng / mL ligand Flt-3, 20 ng / mL IL-7 and 10 ng / mL IL-15.
NK2C medium: RPMI 1640 (Cat # 22400105, Invitrogen) supplemented with 10% FBS (Cat # SH30070.03, Hyclone), 2 mM L-glutamine, 1% pen / strep, 50 ng / mL SCF, 50 ng / mL Ligand Flt-3, 100 IU / mL IL-2, 20 ng / mL IL-7 and 20 ng / mL IL-15.
NK2D medium: serum free medium (StemSpan, Cat # 09650, Stem Cell Technologies, Vancouver, Canada) supplemented with 1 μ? Synthetic glucocorticoid dexamethasone (Dex, Cat # D4902, Sigma, St. Louis, MO), 40 ng / mL insulin-like growth factor 1 (IGF-1, Cat # 291-G1-250, R &D Systems, Minneapolis, MN ), 100 ng / mL SCF, 40 ng / mL lipids (mixture of lipid-rich co-cholesterol, Cat # C7305-1G, Sigma, St Louis, MO), 5 ng / mL IL-3, 200 lU / mL IL-2 , 20 ng / mL IL-7 and 20 ng / mL IL-15.
The harvested cells were washed at different time points twice with TPM11640 (phenol-free) and 5% FBS, labeled with fluorescence-conjugated antibodies (Tables 2 and 3) for 15 minutes at 4 ° C, and analyzed by flow cytometry. (FACSCanto, BD) and flow cytometry software Jo (Tree Star).
Table 2. Antibodies used for cell tag Table 3. NK surface receptor flow cytometric characterization panel Cytotoxicity assay using PKH26 / TO-PRO-3 tag. Target tumor cells were labeled with PKH26 (Cat # PKH26GL, Sigma-Aldrich), a dye that inserts into cellular plasma membrane via its lipophilic lytic residue, then placed in 96-well U-bottom tissue culture plates. incubated with expanded NK cells at various effector-objective relationships (E: T) at 200μ? RPM! 1640 supplemented with 10% FBS. Cultures were incubated for 4 hours at 37 ° C in 5% C02. After incubation, cells were harvested and TO-PRO-3 (Invitrogen Cat # T3605), a DNA stain impermeable to membrane, was added to cultures (1 μm final concentration) followed by FACS analysis. Cytotoxicity was expressed as the percentage of dead cells (PKH26 + TO-PRO-3 +) within the total PKH26 + target tumor cells.
Optimization of expansion and differentiation of CD34 * cells in NK cells. Among the media tested formulations, media NK1, NK2, and NK3, NK1 showed an expansion of 500 folds on day 21 (D21). Neither the NK2 nor NK3 medium maintained cell proliferation or differentiation. More media optimization was performed for NK1 medium and subsequent media were called NK2A, NK2B, NK2C and NK2D,. CD34 + cells cultured with NK2A medium showed an expansion of 10"5-fold on day 55 (D55). Based on the results of fold expansion, differentiation and cytotoxicity of NK1, 2 and 3 medium, the second batch of Formulations of NK2A, NK2B, NK2C and NK2D media were made and in D55, a 105-fold expansion was achieved (as shown in Figure 1.) The NK2B medium showed an expansion of approximately 3 x 10-fold. in NK2C medium resulted in expansion of 3 x 102-fold in 21 days, followed by decreased cell viability.NK2D medium did not maintain cells throughout the duration of the experiment.
On day 48 (D48), about 90% of the NK cells in NK2A medium were CD56 + CD3 ~. Within the population CD56 + CD3", more than 98% of cells were CD56 + CD16" (as shown in Figure 2), while 58% expressed the activating receptor NKG2D, 68% were NKp46 + and 17% were CD226 + ( as shown in tables 4A and 4B).
Table 4A, 4B. Phenotypic characterization of expanded NK cells in D48 Additionally, 97.8% of cells cultured in NK2A medium and 93.1% of cells cultured in NK2B medium were CD56 + CD16"on day 21 of culture. 7. 3 Example 3: Culture of NK Cells in CNK Medium Power Expansion and Cytotoxicity of NK Cells On day 27 (D27), CD34 + cells cultured in the NK2A medium were further cultured in one of the following media: • Two-stage medium, comprising CNK medium and Maintenance Medium. CNK medium is IMDM (Invitrogen) supplemented with 10% FCS (Hyclone), 200 lU / mL IL-2 (R &D Systems), 35 Mg / mL transferin (Sigma-Aldrich), 5 μg / mL insulin (Sigma- Aldrich), 2 x 10'5 M ethanolamine (Sigma-Aldrich), 1 μ9 /? Oleic acid (Sigma-Aldrich), 1 μg / mL linoleic acid (Sigma-Aldrich), 0.2 μg / mL palmitic acid ( Sigma-Aldrich), 2.5 μ9 / β-BSA (Sigma-Aldrich) and 0.1 μg / mL phytohaemagglutinin (PHA-P, Sigma-Aldrich). CD56 + CD3"NK cells cultured in NK2A medium were suspended at 2.5 x 10 5 live cells / mL in CNK medium in 24-well plates treated with cell culture or T-bottles. Allogeneic PBMC treated with mitomycin C and K562 cells (line chronic myelogenous leukemia cell) were added to the medium of CNK as feeder cells, at a final concentration of 1 x 106 per mL. NK cells were cultured for 5-6 days at 37 ° C in 5% C02. After 5-6 days and then every 3-4 days a volume equal to maintenance medium (IMDM with 10% FCS, 2% AB serum) human, antibiotics, L-glutamine and 400 units of IL-2 per ml_) was added to! culture.
• NK2A medium (PDAC) with placental stem cells adherent to tissue culture plastic CD34", CD10 +, CD105 +, CD200 + treated with mitomycin C as feeder cells; • NK2A medium (MSC) with mesenchymal stem cell (MSC) treated with mitomycin C as feeder cells; or · Feeding free NK2A medium (FF) as the control.
The two-stage medium enhanced the fold expansion of CD34 + cells compared to NK2A (FF), NK2A (PDAC) and NK2A (MSC), in particular between day 27 (D27) and day 48 (D48). See figure 3.
By day 35, the proportion of CD34 + cells had already decreased to approximately 4% while the proportion of CD56 + CD3"had increased to approximately 80% in the two-stage medium." Day 45 (D45), the cells cultured in Two-stage media showed higher cytotoxicity compared to NK cells treated in NK2A (FF), NK2A (PDAC) and NK2A (MSC) (as shown in Figure 3) .Phenotypic characterization on day 41 (as shown) in Figure 5) showed increased expression of NKp46 and CD226 in the cells, indicating a possible explanation for the improvement of cytotoxicity.In D41, the proportion of CD226 + cells increased from 0.9% ± 0.8% in NK2A medium to 13% ± 4% in the middle of two stages, the proportion of NKp46 + cells increased from 55.4 ± 8.7% in NK2A medium to 80% ± 7.85% in two-stage medium In D48 the proportion of CD226 + cells increased from 17.3 ± 14.3% in mean of NK2A to 52.3% ± 11.64% in the middle of two stages; NKp46 + cells increased from 67.9% ± 5.4% in NK2A medium to 86% ± 4% in two stages. There was no significant difference in NKG2D expression between the conditions tested. The changes in expression of CD226 and NKp46 are shown in Table 5, below.
Table 5. Expression of CD226 and NKp46 in NK cells cultured in NK2A (FF) and two-stage medium on day 41 (D41) and day 48 (D48). The standard deviation was calculated for 3 donors. 7. 4 Example 4: Comparison of Natural Killer Cells Cultivated in Two Step Medium and Natural Killer Cells Derived from Embryonic Stem Cells (ESCs) NK cells in the two-stage medium were compared with NK cells derived from embryonic stem cells (ESCs), which were produced by the method of Woll et al., Blood 113 (4): 6094-6101 (2009). Specifically, a difference in expression levels of CD94 and CD117 was observed during the culture process of both cell types. Figure 6 shows that CD117 expression was high in two-step NK cells, or "+", from Day 7 (D7) to Day 35 (D35), while CD94 expression increased gradually. At day 35 (D35), approximately 44% of the CD56 + CD3"(two steps) 'cells were CD94 + CD117 \ 37.6% of CD56 + CD3 cells were CD94" CD117 + and 14.7% of CD56 + CD3 cells "They were CD94 + CD117 cells". As such, the NK cells produced by the two-step method are distinguishable from the NK cells derived from ESCs, 78% of which remained CD117baio / "from day 14 to day 35 of the culture.This difference in CD117 expression is already useful. that NK cells CD117 + are cytotoxic towards tumor cell lines of various tissues, as described in Example 6, below.
These results suggest that the progress of differentiation of TSNK cells is different from NK cells derived from ESC, and that TSNK cells are distinguishable from NK cells derived from ESC. 7. 5 Example 5: PDACs Improve Fold Expansion of Natural Killer Cells Cultivated in NK2A Medium To evaluate the effect of placental stem cells adherent to tissue culture plastic CD10 +, CD34", CD105 +, CD200 + (referred in this example as PDACs) in differentiation of hematopoietic stem cell (HSC) to natural killer cells, HSCs were stimulated with PDACs treated with mitomycin C or mesenchymal stem cells derived from bone marrow (MSC) at a ratio of 10: 1 PDACs / MSC: HSC on day 0 and day 21, while a feeder-free culture was used as the control NK2A medium was used as a culture medium PDACs were found to improve the expansion of fold of cultured NK cells compared to medium alone, however, no significant difference in cytotoxicity was found between cells grown with or without the feeder layer.The cells treated with MSC showed the highest fold expansion, but the lowest cytotoxicity , as shown in figure 7. 7. 6 Example 6: Cytotoxic Activity of Expanded NK Cells Using Two Stage Media This example demonstrates that the NK cells produced from CD34 + cells expanded and differentiated using the two-step process described above are cytotoxic to tumor cell lines.
Dehydrogenase release test of lactate (LDH). The LDH release assay was performed using CYTOTOX 96® non-radioactive cytotoxicity equipment (Promega, Cat # G1780). In this assay, cultured NK cells derived from donor-compatible human placenta perfusate (HPP) and cord blood units (Combos units) were used as effector cells, while certain tumor cell line cells were used as target cells. From three units in this study, the percentage of HPP cells was 56.6 ± 28.3%. Effector cells and target cells were placed in 96-well U-bottom tissue culture plates and incubated at various effector-target (E: T) ratios at 100 μ? RPMI 1640 without red phenol (Invitrogen, Cat # 11835-030) supplemented with 2% human serum AB (Gemini, Cat # 100-512). Cultures were incubated for 4 hours at 37 ° C in 5% C02. After incubation, 50 μ? of supernatant was transferred to enzyme assay plate, LDH activity was detected as provided by the manufacturer, and absorption was measured at 490 nm in an ELISA reader (Synergy HT, Biotek). The cytotoxicity was calculated according to the following equation:% Cytotoxicity = (Sample = Spontaneous Efectora - Spontaneous Diana) / (Maximum Diana - Spontaneous Diana) * 100, where "Spontaneous Efectora" is a control for the spontaneous release of LDH from effector cells; "Spontaneous Diana" is a control for the spontaneous release of LDH from target cells; and "Diana Maximum" is a control for maximum LDH release when essentially 100% of the cells are lysed.
Isolation of CD34 + Human Placenta Perfused Cell (HPP) and CD34 + Umbilical Cord Blood Cell (UCB). HPP and UCB cells were purified using Ficoll or ammonium chloride to obtain total nucleated cells (TNCs). TNCs were then used in a positive selection procedure to isolate CD34 + cells using anti-CD34 and RoboSep spheres following the protocol provided by the manufacturer (StemCell Technologies, Inc.). In this experiment, CD34 + cells with more than 90% purity were isolated.
Tumor cell susceptibility study to two-stage NK cells cultured. Tumor cell lines (table 1), including human breast cancer (HCC2218), human colorectal adenocarcinoma (HT-29), human chronic myelogenous leukemia (CML), human acute myeloid leukemia (AML), human glioblastoma (LN-18 and U-118MG), human multiple myeloma (U266), human histiocytic lymphoma (U937), and human retinoblastoma (WERI-RB-1) were co-cultured in two-stage NK cells. NK cells cultured in two stages included those cultured in NK2A medium for 21 days, then cultured in CNK medium for 21 days, and those cultivated in NK2A medium for 28 days and then CNK medium for 14 days. The NK cell cytotoxicity was measured by the lactate dehydrogenase (LDH) release assay after 4 hours co-culture. At an effector to target ratio (E: T) of 10: 1, the latter usually showed greater cytotoxicity than the first (Table 6). Of the tumor cell lines, LN-18 was the most susceptible to killing mediated with NK, followed by K562, U937, WERI-RB-1, U-118MG, HT-29, HCC2218, KG-1 and U266.
Therefore, TSNK cells showed significant cytotoxicity towards several cancer cell lines. Furthermore, it seems that the NK cytotoxicity can be improved by prolonging the culture period in NK2A medium from 21 days to 28 days.
Table 6. Cytotoxicity of cultured TSNK cells focusing on tumor cell lines MicroRNA profile of Human Placenta Perfused CD34 + Cells (HPP) and CD34 + Umbilical Cord Blood Cells (UCB). Purified donor-compatible HPP and CD34 + cells were subjected to microRNA (miRNA) preparation using a miRNA IRVANA ™ isolation kit (Ambio, Cat # 1560). CD34 + cells (0.5 X 10 6 cells) were disrupted in a denaturing lysis regulator. The samples were then subjected to acid-phenol + chloroform extraction to isolate highly enriched RNA for small RNA species. 100% ethanol was added to take the samples to 25% ethanol. When this mixture of lysate / ethanol passed through a glass fiber filter, large RNA species were immobilized, and the small RNA species were collected in the filtrate. The ethanol concentration of the filtrate was then increased to 55%, and the mixture passed through a second glass fiber filter where the small RNAs are immobilized. This RNA was washed few times, and eluted in a low resistance ionic solution. The concentration and purity of the recovered small RNA was determined by measuring its absorbance at 260 and 280 nm. It was discovered that miRNAs were unique for CD34 + HPP cells in all tested donors (n = 3) including hsa-miR-380, hsa-miR-512, hsa-miR-517, hsa-mR-518c, hsa-miR-519b, and hsa-miR-520a. 7. 7 Example 7: Isolation of CD34 * cells from UCB and HPP pooled This example demonstrates the isolation of CD34 + cells from umbilical cord blood (UCB) and perfused human placenta (PPH) grouped (Combo). To evaluate the grouping relationship of UCB: HPP, side-by-side comparisons were made from 3 different grouping ratios as follows: (1) full grouping: 1x UCB (full volume) + 1x HPP (full volume); (2) partial grouping of HPP (33%): 1x UCB (full volume) + 0.33x HPP (1/3 volume HPP); and (3) partial grouping of HPP (10%): 1x UCB (full volume) + 0.10x HPP (1/10 volume HPP). A total of N = 3 experimental replicas was executed. Initial TNC and volumes were recorded. The pooled samples were then purified for CD34 + cells and CD34 + purity was determined post-thawing. The optimal grouping ratio was then determined graphically of the purity CD34 + post-thawing vs. Volumetric fractions or cell count (TNC) (as combo%) graphs. As shown in Figure 8, the endpoint CD34 + purity correlates well with the volume content of HPP, but not so much with the HPP TNC content. Overall, UCB 85% v / v, HPP 15% v / v was found to be the optimal grouping ratio to obtain CD34 + cells with purity of 80% and more. 7. 8 Example 8: Comparison of NK Cell Culture Using medium based on GBGM® This example demonstrates the comparison of NK cell culture processes using two GBGM-based media (the three-step process and the two-step process). The two processes are summarized in Table 10. Both processes used GBGM as the basal medium for the differentiation of NK cells and CD34 + cells from placenta in origin.
Table 7. Summary of the processes of three stages and two stages The experimental parameters are outlined below: Lots of donors: (1) CD34 + cells from fresh UCB: N = 6 (2) Cool Combo CD34 + cells: N = 2 (3) Cryoconserved "Combo" CD34 +: N = 8 Scale: Multi-bowl dishes to T-25, up to multiple bottles T-75, mL in culture volume Process Methods: (1) Two stage process (2) Three stage process Use of feeders: (1) Without feeders (2) With K562 & Inactivated PBMC, added to culture on day 21 Seedbed density: 20000 - 50000 cells / mL CD34 + cells from fresh UCB or fresh combo were generated and cryopreserved with methods described in Examples 7, 10 and 11. Cultures were maintained in an incubator at 37 ° C, > 90% humidity and 5% C02. Cell growth was monitored (cell count) and media exchanges were made twice a week to maintain cell concentrations within the range of 5 x 104 - 1 x 106 per ml_. The differentiation was monitored by phenotypic analysis on day 21 and 35. When feeders were used, on day 21 of NK culture, fresh 3-day cultivated K562 and post-thawing allo-PBMC with mitomycin-C (16 μg) were inactivated. mL> 2 hours, 37 ° C) and were added to the two stage process conditions at 1 x 106 per mL. On day 35, after performing final cell counts, a cytotoxicity assay based on flow cytometry using K562 cells labeled with PKH and freshly cultured (ratio E: T 10: 1, 4 hr, 37 ° C) was performed to evaluate NK functionality.
Results The mean of the TNC expansion fold for CD34 + cells derived from fresh UCB using the two processes was comparable at day 35. The two-step process appeared to produce more TNC expansion fold than the three-step process for CD34 + cells derived from cool combo The TNC global expansion folds were comparable for CD34 + cells derived from post-thawing combo using both processes, but were significantly lower than those derived from fresh UCB or fresh combo. In all, both two-stage and three-stage processes produced similar cellular performance at the end of the culture.
On day 21, there were no notable phenotype differences in cells originating from UCB or Combo. The two-step process resulted in a higher percentage of CD56 + CD3 NK cells (average 14.7%) than the three-stage process (average 6.1%) .The degree of differentiation (e.g., CD56 + CD3 ~ level) be dependent on donor The percentage of both populations CD3 + CD56"(T cells) and CD3 + CD56 + (NKT type cells) were minimal in all cases.
On day 35, it was discovered that both processes are effective in differentiating NK cells, as evidenced by the CD56 + CD3 purity levels of "high endpoint (87.2% and 90.1% for the two-stage and three-stage processes respectively The addition of feeders in the two-stage process appeared to improve the phenotypic purity of NK (75.3% without feeders, 87.2% with feeders), although no observable benefit of feeders was found on NK purity (90.1% without feeders; % with feeders) with the three-stage process.
Cultured NK cells maintained their production of CD16 'phenotype. The two-step process appeared to produce slightly more CD56 + CD3 + cells in the absence of feeders (average 11.2%) than the other conditions (< 2%). The presence of PBMC and K562 feeders in both processes significantly up-regulated / activated certain functional NK markers (NKp46, DNAM-1, CD94) in the NK cell population. Overall, the purity expression profiles of NK and functional marker were found to be compatible between the two processes, when the feeder conditions were identical.
The functionality of cultured NK cells, as determined by the in vitro 4-hour K562 cytotoxicity assay, was found to be comparable between the two processes when the feeder conditions were identical. The NK cells activated with feeder were highly effective in killer K562 cells in vitro, with average specific lysis of 93.2% and 93.6% specific lysis for the two-stage and three-stage processes, respectively.
In summary, the two-step and three-step processes were found to produce comparable growth, phenotype (purity and activation markers), and in vitro functionality for NK cells when the same feeder condition was used. The two-stage process offers the ease and convenience of growing NK cells compared to the three-stage process. 7. 9. Example 9: Comparison of NK Cell Culture Using Various Basal Media This study seeks to evaluate the differentiation and expansion of NK cells derived from CD34 + using different basal media.
The experimental conditions are summarized in Table 11. Cells were cultured as described in Example 11. All experiments were placed on the scale of multi-well dishes / T-flasks and were maintained at 37 ° C, > 90% humidity, and 5% C02 incubator. Cell growth was monitored (cell count) and media exchanges were made twice a week to maintain cell concentrations within the range of 5x104 - 1x106 per mL. The differentiation was monitored by phenotypic analysis on day 21 and day 35. On day 21, K562 cultured in 3 days fresh and post-thawing allo-PBMC were inactivated and added to growing NK cell culture at 1x106 per mL. The NK cells were normalized to 0.5x106 per mL. On day 35, after final cell counts were performed, a cytotoxicity assay based on flow cytometry using K562 cells labeled with PKH and freshly cultured (ratio E: T 10: 1, 4 hr, 37 ° C) was carried out to evaluate the functionality of NK.
Table 8. Summary of experimental conditions for evaluation of several basal media Growth Performance Stemspan H3000 and OpTimizer showed comparable growth performance (TNC expansion fold) to GBGM at day 35. Cellgro, X-VIVO 15, AIM-V, X-VIVO 10, DMEM: F12, DMEM: F12 the 5 mM OAC showed growth performance lower than GBGM.
Phenotypic analysis On day 35 (endpoint), GBGM produced approximately 80% purity of CD56 + CD3 cells. "OpTimizer and Stemspan H3000 produced approximately 50% CD56 + CD3 + DMEM purity: F12 produced approximately 35% CD56 + CD3 purity AIM-V, X-Vivo 10, X-VIVO 15 and half Cellgro produced approximately <30% purity of CD56 + CD3 'cells. The addition of OAC to DMEM: F12 basal medium during culture was found to greatly enhance the end-point NK purity, the percentage of CD56 + CD3 'on day 35 of the culture increased from 35% to 72%.
Cito toxicity / Functionality The addition of 5 mM OAC to DMEM: F12 basal medium during culture was found to greatly enhance the activation status and in vitro functionality of NK cells. The addition of OAC also significantly increased the level of NK activation markers NKp46, NKG2D, DNAM-1 and CD94. The in vitro functionality (K562 cytotoxicity) of day 35 NK cells also increased substantially, from 21.4% to 97.1%.
Overall, the NK cell properties improved significantly from the addition of 5 mM OAC. 7. 10. Example 10: Storage and Cryoconservation of NK Cells This example demonstrates the methods of storing and cryopreservating NK cells. The CD34 + hematopoietic stem cells isolated from human placental perfusate (HPP) and umbilical cord blood cells were expanded and differentiated into NK cells using the protocols described in the previous examples. The cells were cryopreserved immediately after being isolated from HPP and umbilical cord blood (on day 0) or during the first growth phase of NK cells (on day 9, day 14, day 21 or day 35 post isolation).
The cells were cryopreserved in the following cryopreservation formulations: Formulation 1 - dextran cryo medium: 5% DMSO (Sigma Aldrich, D2650), 55% dextran (10% w / v in normal saline) (10% LMD in 0.9% sodium chloride injection, Hospira), 40% HAS (Octapharma); Formulation 2 - half of trehalose cryo: 5% DMSO, 55% trehalose (10% w / v in normal saline), 40% HSA; Formulation 3 - CryoStor® CS2 (BioLife Solutions); Formulation 4 - CryoStor® CS5 (BioLife Solutions); Formulation 5 - CryoStor® CS10 (BioLife Solutions); Formulation 6 - free serum freezing medium (Sigma-Aldrich, Cat # 6295); Formulation 7 - Glycerol freezing medium (Sigma-Aldrich, Cat # C6039); or Formulation 8 - DMSO and free serum freezing medium (Sigma-Aldrich, CAT # 2639).
Cells harvested at different time points were washed several times with culture medium or saline. The cells were then centrifuged to obtain cell pellets. The supernatants were removed, and the cell pellets were suspended with cryopreservation medium at approximately 1 x 106-1.5 x 10 7 or more cells per milliliter. The cell suspension was aliquoted to 1mL or 2mL septum flasks and incubated at 2-8 ° C for approximately 10 minutes. Subsequently, the cells were frozen in a control speed freezer (Thermo) to 0. 5 ° C / min. Frozen flasks were transferred to a cryogenic freezer for vapor storage of liquid nitrogen. The cryopreserved NK cells were thawed rapidly in a 37 ° C water bath with careful turbulence of the samples until all the visible ice melted. The cell samples can be diluted with pre-heated culture medium. 7. 11. Example 11: Storage and Cryoconservation of NK Cells This example demonstrates another method of storing and cryopreservating NK cells. The CD34 + hematopoietic stem cells isolated from human placental perfusate (HPP) and umbilical cord blood cells were expanded and differentiated into NK cells using the protocols described above. Cells were cryopreserved immediately after being isolated from HPP and umbilical cord blood (on day 0) or during the first growth phase of NK cells (on day 9, day 14, day 21 or day 35 after isolation).
A cell suspension solution was prepared by combining Dextran-40 and HSA in the ratio of 60% Dextran-40 v / v, 40% HSA (from a 25% solution) v / v).
A solution of 2x thawing was prepared with 50% Dextran-40 v / v, 40% HSA (25% solution) v / v, 10% DIVISO v / v. DMSO was first added slowly to Dextran-40 and mixed well.
Subsequently, 25% HSA solution was added to the solution slowly with mixing. The resulting solution was mixed well and brought to room temperature before use.
Cryopreservation procedure The cell number was estimated and normalized as a cell suspension at 15x106 in cell suspension solution. The volume of the cell suspension was determined and an equal volume of freshly prepared 2x freezing solution was recorded and distributed to a number of bottles. MycoAlert test was performed in culture supernatants saved to detect mycoplasma contamination. A post-thaw test was performed in a retention bottle to determine post-thaw viability, cell recovery and cell characterization. 7. 12. Example 12: Analysis of cryopreserved / thawed NK cells Feasibility test. The NK cells cryopreserved in various formulations as in example 10 or 11 were thawed. Cells were frozen at the density of 2 x 106-3 x 107 cells / mL. The thawed NK cells were evaluated for cell viability using the Countess® automated cell counter (Invitrogen) on day 0, day 3 and day 18 after thawing compared to fresh cells or pre-frozen cells. Briefly, 10 μ? of cell samples were mixed with 10 μ? of blue triptan. The cell mixtures were pipetted into the Countess® camera slide. The slide was inserted into the instrument and the cells were counted. Post-freeze-thaw cells showed cell viability approximately 80% at > 90% viability, depending on different formulations of cryopreservation.
Apoptosis trial. The thawed NK cells were also evaluated for apoptosis using BD AnnV / PI Apoptosis assay kit on day 0, day 3 and day 18 after thawing. Briefly, cells were washed twice with cold 1x PBS and re-suspended in 1x binding buffer (BD Annexin V / PI kit Apoptosis part number 556547 Bonding composition regulator part number 51-66121 E 0.1M Hepes / NaOH (pH 7.4), 1.4M NaCl, 25m CaCl2 For 2x diluted 1 part 10x regulator to 9 parts of distilled water). 100μ? of cell suspension containing approximately 100,000 cells was transferred in a FACS tube. 100μ? of 1x union regulator, 5μ? of Ann V-FITC, and 5μ? of PI-PE was added to the tube. The tube was then mixed in a vortex carefully and incubated in the dark for 15 minutes. Subsequently, 400 μ? of 1x binding regulator was added. The samples were analyzed in 1 hour. The controls used to establish quadrants and grids were non-stained cells, cells stained with Ann V-FITC only and not Pl, cells stained with Pl only and not AnnV. The apoptotic cells were quantified as% of the population of lattice cases as "cells" in the size dispersion chart (FSC vs SSC). Post-freeze-thaw cells showed approximately 5-25% of dead / late apoptotic cells and 10-25% of early apoptotic cells, depending on different cryopreservation formulations. Overall, formulation 1 (5% DMSO, 55% dextran (10% w / v in normal saline), 40% HSA), formulation 2 (5% DMSO, 55% trehalose (10% w / v in saline) normal), 40% HSA), formulation 4 - CryoStor® CS5 (BioLife Solutions), and formulation 5 (CryoStor® CS10 (BioLife Solutions) showed greater cell viability and fewer apoptotic cells compared to other formulations. 7. 13. Example 13: Evaluation of Cryopreserved Cellular Banking Process This example demonstrates the evaluation of In-Process Cryoconserved Cellular Banking. Cell culture was started with UCB CD34 + cells using the method as described by Spanholtz et al., PLoS One 5 (2): e9221 (2010) using HS-AB or FBS as the source of serum. The cell concentration was determined and adjusted, and the medium was replenished as necessary. On day 7, 9, 10 or 14, approximately 106-3 x 106 cells were removed from the cell culture, centrifuged and resuspended in cryopreservation medium (5.5% v / v Dextran-40, 10% v / v HSA, 5% v / v DMSO). The cells were frozen in a controlled speed freezer and transferred to liquid phase nitrogen storage for cryopreservation. Approximately 1ml_ cells per bottle were cryopreserved at a concentration ranging from 106 - 107 per ml_. The remaining crop was taken to the final point (day 35), referred to as "fresh (no cryopreservation in process)". Phenotypic analyzes were performed on day 21, 28 and 35 of the cell culture. The in vitro functionality (cytotoxicity K562, E: T 10: 1) was evaluated at day 35 (end point) of the culture.
The post-thawing operation of the cryopreserved culture samples in process (day 9 and 14) were cultured in the following manner. The cell bank flasks were thawed rapidly in the 37 ° C water bath. The cells were then diluted with RPMI-FBS medium, centrifuged, resuspended and seeded in culture medium. Each of the cultivation conditions was carried forward to day 35, cumulative from the beginning of the cultivation process. Analytics (cell counting, viability, phenotypic analysis, functionality evaluation) were done in the same way as their "fresh" counterparts.
Results Cryopreserved samples in process of day 9, 10 and 14 all produced excellent post-thawing viability, regardless of the point of time in process or cellular concentration (96.2% -97.3% blue triptan negative, 86.1% -93.2% Annexin-V negative / TO-PRO 3 negative). The banking in process had no negative effects on loss of final crop yield (day 35). The final cellular yield between day 9 or day 14 post-thawing and "fresh" conditions are comparable, with HS-AB or HS-AB as the source of serum.
The phenotypic profiles of NK cells maturing at day time points 21/28/35 were quite comparable between the "fresh" and "post-thaw" cultures, respectively. The phenotypic purity (CD56 + CD3) was comparable as well. The expression of certain functional NK markers (CD94, NKG2D) were slightly different due to variabilities.
In the K562 cytotoxicity assay, post-thawing NK cells grown on day 9/14 were found to produce -0-20% lower specific K562 lysis reading compared to non-processed cryopreserved NK cells. However, given the expression levels of surface markers relevant to NK cytotoxic function (DNAM1, NKp46, NKG2D, etc.) were not reduced concurrently, the trend would need to be confirmed with additional donors and test repetitions. Overall, cryopreservation in process on day 9/14 of cultivation had minimal impact on the outcome of the process. 7. 14. Example 14: Development of post-thawing medium for NK cell dosing This example demonstrates the development of post-thaw medium for NK cell dosing in animals. The effects of injection medium and cell density were tested on NK cell viability, cytotoxicity, cell recovery and lump formation. The viability was evaluated by spotting with tryptan blue; The cytotoxicity was evaluated by FACS (10: 1 ratio of NK: K562) and the formulation of the lump was evaluated by microscopic assay. The results are shown in tables 9-12 for the various types of injection medium and cell density.
Table T. Cell recovery, viability, cytotoxicity and lump formation of specific conditions tested Table 10. Cell recovery, viability, cytotoxicity and lump formation of specific conditions tested Table 11. Cell recovery, viability, cytotoxicity and lump formation of specific conditions tested Table 12. Cell recovery, viability, cytotoxicity and lump formation of specific conditions tested The results showed that the injection medium of Plasmalite + 1% HSA maintained NK cells with better viability and cytotoxicity than PBS injection medium + 1% FBS. The cytotoxicity also decreased over time after suspending the cells in injection medium. There is no effect of cell density observed in viability and cytotoxicity when the cells were suspended in Plasmalita + 1% HSA. The NK cells were also established in PBS + 1% FBS or Plasmalite + 1% HSA medium after 1 hour; however, the cells did not appear to be added. Finally, there was no obvious loss of cellular recovery and viability observed from the freeze-thaw process. 7. 15. Example 16: Development of Post-deconditioning medium for NK cell dosing The effects of various concentrations of HSA were also tested on NK cell viability, cytotoxicity, cell recovery and lump formation. The same methods were used from Example 19. The results are shown in Tables 14-16 for the various types of injection medium and cell density.
Table 13. Cell recovery, viability, cytotoxicity and lump formation of specific conditions tested Table 14. Cell recovery, viability, cytotoxicity and lump formation of specific conditions tested Table 15. Cell recovery, viability, cytotoxicity grouping of specific conditions tested Table 16. Cell recovery, viability, cytotoxicity and lump formation of specific conditions tested The results show that the viability remained well over 4 hours post-thawing in the three injection media tested. Also, the cytotoxicity decreased with time in three injection media. However, the level of reduction was lower in higher concentrations of HSA while Plasmalita + 5% HSA maintained the highest cytotoxicity. It was also observed that NK cells do not aggregate in all three injection media. Overall, Plasmalite appears to be a better candidate for injection medium than PBS. 7. 16. Example 17: Culture of NK cells without IL-2 This example demonstrates the culture of NK cells in the absence of IL-2. Cell culture was performed by the two-step process described in Example 11. Five different concentrations of IL-2 in the first medium were tested: 0, 200, 500, 1000, 2000 U / mL.
The results indicate that developing NK cells in culture did not appear to respond to growing IL-2. The purity of NK cells did not seem to depend on IL-2; NK cells differentiate into CD56 + CD3- phenotype in the absence of IL-2. The combination of IL-7, IL-15 and SCF appeared to be sufficient for cell development NK in vi tro.
Equivalents: The present invention should not be limited in scope by the specific embodiments described herein. In fact, various modifications of the invention in addition to those described will be apparent to those skilled in the art from the following description and accompanying figures. Such modifications should fall within the scope of the appended claims.
All references cited herein are incorporated herein by reference in their entirety and for all purposes to the same degree as if each individual publication, patent or patent application individually and individually was indicated for be incorporated by reference in its entirety for all purposes. The citation of any publication is for description before the filing date and should not be considered an admission that the present invention should not precede said publication by virtue of the prior invention.

Claims (25)

1. - A method of producing a population of natural killer cells (NK), comprising: (a) planting a population of hematopoietic stem or progenitor cells in a first medium comprising interleukin-15 (IL-15) and, optionally, one or more of the stem cell factor (SCF) and interleukin-7 (IL-7) , wherein said optional IL-15 and SCF and IL-7 are not comprised within an undefined component of said medium, such that the population expands, and a plurality of hematopoietic stem or progenitor cells within said population of cells hematopoietic stem or progenitors differentiate into NK cells during said expansion; and (b) expanding the cells of step (a) in a second medium comprising interleukin-2 (IL-2), to produce a population of activated NK cells.
2. A two-step method of producing a population of natural killer (NK) cells, wherein a first step of said method comprises expanding a population of hematopoietic stem or progenitor cells into a first medium comprising one or more of the stem cell factor (SCF), interleukin-7 (IL-7) and interleukin-15 (IL-15), and wherein said SCF, IL-7 and IL-15 are not comprised within an undefined component of said medium, and in where a plurality of hematopoietic stem or progenitor cells within said population of stem cells or Hematopoietic progenitors differentiate into NK cells during said expansion; Y wherein a second step of said method comprises expanding the cells from the first passage into a second medium comprising interleukin-2 (IL-2), to produce activated NK cells.
3. - The method according to claim 1, wherein the first medium further comprises one or more of tyrosine kinase 3 ligand type Fms (Flt3-L), thrombopoietin (Tpo), interleukin-2 (IL-2) or heparin.
4. - The method according to claim 3, wherein the first medium further comprises fetal bovine serum or human serum.
5. - The method according to claim 3, wherein the SCF is present at a concentration of about 1 to about 150 ng / mL in the first medium.
6. The method according to claim 3, wherein the Flt3-L is present at a concentration of about 1 to about 150 ng / mL in the first medium.
7. The method according to claim 3, wherein the IL-2 is present at a concentration of about 50 to about 1500 IU / mL in the first medium.
8. The method according to claim 3, wherein the IL-7 is present at a concentration of about 1 to about 150 ng / mL in the first medium.
9. The method according to claim 3, wherein the IL-15 is present at a concentration of 1 to about 150 ng / mL in the first medium.
10. The method according to claim 3, wherein the IL-15 is present at a concentration of 1 to about 150 ng / mL in the first medium.
11. - The method according to claim 3, wherein the TPO is present at a concentration of about 1 to about 150 ng / mL in the first medium.
12 -. 12 - The method according to claim 1, wherein said IL-2 in the second step is present at a concentration of 50 to about 1500 IU / mL in the second medium.
13. - The method according to claim 1, wherein said second means further comprises one or more of fetal calf serum (FCS), transferin, insulin, ethanolamine, oleic acid, linoleic acid, palmitic acid, bovine serum albumin (BSA) ) and phytohemagglutinin.
14. - The method according to claim 1, wherein the hematopoietic stem or progenitor cells are CD34 +.
15. The method according to claim 1, wherein the haematopoietic stem or progenitor cells comprise hematopoietic stem cells or progenitors of human placenta perfusate and umbilical cord hematopoietic progenitor or stem cells, wherein said placental perfusate and said umbilical cord are from the same placenta.
16. The method according to claim 1, wherein the feeder cells in step (b) comprise peripheral blood mononuclear cells treated with mitomycin C (PBMC), K562 cells or tissue culture adherent stem cells.
17. - The method according to claim 1, wherein the NK cells are CD3"CD56 + CD16".
18. - The method according to claim 17, wherein the NK cells are additionally CD94 + CD117 +.
19. - The method according to claim 17, wherein the NK cells are additionally CD161".
20. - The method according to claim 17, wherein the NK cells are additionally NKG2D +.
21. - The method according to claim 17, wherein the NK cells are additionally NKp46 +.
22. - The method according to claim 17, wherein the NK cells are additionally CD226 +.
23. - A population of activated NK cells obtained by a method comprising: (a) planting a population of stem cells or hematopoietic progenitors in a first medium comprising interleukin-15 (IL-15) and, optionally, one or more of the stem cell factor (SCF) and interleukin-7 (IL-7) , wherein said optional IL-15 and SCF and IL-7 are not comprised within an undefined component of said medium, such that the population expands, and a plurality of hematopoietic stem or progenitor cells within said population of cells hematopoietic stem or progenitors differentiate into NK cells during said expansion; Y (b) expanding the cells of step (a) in a second medium comprising interleukin-2 (IL-2), to produce a population of activated NK cells.
24. A method of suppressing the proliferation of tumor cells comprising contacting the tumor cells with a plurality of NK CD94 + CD117 cells wherein said NK cells have been produced by the method according to claim.
25. The method according to claim 24, wherein said tumor cells are primary ductal carcinoma cells, glioblastoma cells, leukemia cells, acute T cell leukemia cells, chronic myeloid lymphoma (CML) cells, leukemia cells acute myelogenous, chronic myelogenous leukemia (CML) cells, lung carcinoma cells, colon adenocarcinoma cells, histiocytic lymphoma cells, multiple myeloma cells, colorectal carcinoma cells, colorectal adenocarcinoma cells, prostate cancer cells or cells of retnoblastoma.
MX2013000395A 2010-07-13 2011-07-13 Methods of generating natural killer cells. MX342995B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US36398110P 2010-07-13 2010-07-13
US201161497897P 2011-06-16 2011-06-16
PCT/US2011/043831 WO2012009422A1 (en) 2010-07-13 2011-07-13 Methods of generating natural killer cells

Publications (2)

Publication Number Publication Date
MX2013000395A true MX2013000395A (en) 2013-03-22
MX342995B MX342995B (en) 2016-10-21

Family

ID=44546146

Family Applications (1)

Application Number Title Priority Date Filing Date
MX2013000395A MX342995B (en) 2010-07-13 2011-07-13 Methods of generating natural killer cells.

Country Status (13)

Country Link
US (5) US8926964B2 (en)
EP (2) EP3358007A1 (en)
JP (5) JP5996533B2 (en)
KR (7) KR20130093091A (en)
CN (3) CN107828727A (en)
AU (1) AU2011279201B2 (en)
BR (1) BR112013000822B1 (en)
CA (1) CA2804750C (en)
ES (1) ES2666746T3 (en)
IL (2) IL224204B (en)
MX (1) MX342995B (en)
NZ (2) NZ605505A (en)
WO (1) WO2012009422A1 (en)

Families Citing this family (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100915482B1 (en) * 2000-12-06 2009-09-03 하리리 로버트 제이 Method of collecting placental stem cells
NZ528035A (en) 2001-02-14 2005-07-29 Robert J Hariri Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
PL2471904T3 (en) 2005-12-29 2019-08-30 Celularity, Inc. Placental stem cell populations
CA2850793A1 (en) 2006-10-23 2008-05-02 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
EP2129775A1 (en) * 2007-02-12 2009-12-09 Anthrogenesis Corporation Hepatocytes and chondrocytes from adherent placental stem cells; and cd34+, cd45- placental stem cell-enriched cell populations
US9200253B1 (en) 2007-08-06 2015-12-01 Anthrogenesis Corporation Method of producing erythrocytes
EP2783692B1 (en) 2007-09-28 2019-01-02 Celularity, Inc. Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
MX2011001990A (en) 2008-08-20 2011-03-29 Anthrogenesis Corp Improved cell composition and methods of making the same.
CN102176919A (en) 2008-08-22 2011-09-07 人类起源公司 Methods and compositions for treatment of bone defects with placental cell populations
CA2767014C (en) * 2009-07-02 2022-01-25 Anthrogenesis Corporation Method of producing erythrocytes without feeder cells
ES2646750T3 (en) 2010-01-26 2017-12-15 Anthrogenesis Corporation Treatment of bone-related cancers using placental stem cells
SI2556145T1 (en) 2010-04-07 2017-01-31 Anthrogenesis Corporation Angiogenesis using placental stem cells
KR20130092394A (en) 2010-04-08 2013-08-20 안트로제네시스 코포레이션 Treatment of sarcoidosis using placental stem cells
NZ605505A (en) 2010-07-13 2015-02-27 Anthrogenesis Corp Methods of generating natural killer cells
AR093183A1 (en) 2010-12-31 2015-05-27 Anthrogenesis Corp INCREASE IN THE POWER OF PLACENTA MOTHER CELLS USING MODULATING RNA MOLECULES
EP3443968A1 (en) 2011-06-01 2019-02-20 Celularity, Inc. Treatment of pain using placental stem cells
US9925221B2 (en) 2011-09-09 2018-03-27 Celularity, Inc. Treatment of amyotrophic lateral sclerosis using placental stem cells
JP5792886B2 (en) * 2012-03-09 2015-10-14 株式会社日立ハイテクノロジーズ Sample observation method and sample pretreatment method
CN103372029A (en) * 2012-04-19 2013-10-30 孙勇 NK (Natural Killer) cell new technology for treating tumor
EP3722416A1 (en) * 2012-08-13 2020-10-14 Celularity, Inc. Natural killer cells and uses thereof
CN102994449A (en) * 2012-12-13 2013-03-27 上海柯莱逊生物技术有限公司 Method for in-vitro amplification of NK cells
AU2013204922B2 (en) 2012-12-20 2015-05-14 Celgene Corporation Chimeric antigen receptors
WO2014117121A1 (en) 2013-01-28 2014-07-31 St. Jude Children's Research Hospital, Inc. A chimeric receptor with nkg2d specificity for use in cell therapy against cancer and infectious disease
AU2014215458A1 (en) 2013-02-05 2015-08-13 Anthrogenesis Corporation Natural killer cells from placenta
JP5511039B1 (en) * 2013-05-22 2014-06-04 国立大学法人九州大学 Method for preparing NK cells
AU2014348454A1 (en) * 2013-11-15 2016-06-02 Celularity Inc. Compositions comprising human placental perfusate cells, subpopulations thereof, and their uses
JP6775426B2 (en) * 2014-03-07 2020-10-28 エメルセル エスエーエス Pool NK cells derived from cord blood and these uses for the treatment of cancer and chronic infections
EP3143134B1 (en) 2014-05-15 2020-10-28 National University of Singapore Modified natural killer cells and uses thereof
CN105219710B (en) * 2014-06-05 2020-01-10 上海厚超生物科技有限公司 Method for culturing immune cell population with high killing activity
WO2016007827A1 (en) 2014-07-11 2016-01-14 Anthrogenesis Corporation Methods of improving vector transduction efficiency into t lymphocytes
EA201791443A1 (en) * 2014-12-31 2018-01-31 Антродженезис Корпорейшн NATURAL KILLER CELLS AND THEIR APPLICATIONS
WO2016122014A1 (en) * 2015-01-27 2016-08-04 한국생명공학연구원 Method for mass-producing natural killer cells and use of natural killer cells obtained by the method as anticancer agent
TW201718851A (en) * 2015-09-18 2017-06-01 通用醫院公司 Modified natural killer cells having ANTI-FUGETACTIC properties and uses thereof
CN105154400B (en) * 2015-09-30 2021-02-02 中国人民解放军第三0二医院 Amplification method of polyclonal anti-hepatitis B virus immune cells
US10975149B2 (en) 2015-12-16 2021-04-13 The Walter And Eliza Hall Institute Of Medical Research Inhibition of cytokine-induced SH2 protein in NK cells
CA3013187C (en) * 2016-02-01 2023-02-28 Green Cross Lab Cell Corporation Medium composition for cryopreservation of cell and use thereof
CN107267454A (en) * 2016-04-07 2017-10-20 北京京蒙高科干细胞技术有限公司 The amplification in vitro method and its kit of a kind of Cord Blood Natural Killer Cells: Impact and application
CN105886470A (en) * 2016-06-27 2016-08-24 江苏蒙彼利生物科技有限公司 Method for amplifying NK cell
EP3510145A4 (en) 2016-09-06 2020-03-25 The Children's Medical Center Corporation Immune cells derived from induced pluripotent stem cell
CA3056406A1 (en) 2017-03-15 2018-09-20 Orca Biosystems Inc. Compositions and methods for hematopoietic stem cell transplants
WO2018183385A1 (en) 2017-03-27 2018-10-04 National University Of Singapore Truncated nkg2d chimeric receptors and uses thereof in natural killer cell immunotherapy
EP3601532A1 (en) * 2017-03-27 2020-02-05 INSERM (Institut National de la Santé et de la Recherche Médicale) New method to obtain lymphoid progenitors
SG11201908337VA (en) 2017-03-27 2019-10-30 Nat Univ Singapore Stimulatory cell lines for ex vivo expansion and activation of natural killer cells
KR101960410B1 (en) 2017-07-08 2019-03-20 허갑용 Method for preparing metastatic hybrid cells with high fusion efficiency using hypoxic environment induced by cobalt chloride
WO2019046815A1 (en) * 2017-08-31 2019-03-07 Poseida Therapeutics, Inc. Transposon system and methods of use
US20200246393A1 (en) 2017-09-28 2020-08-06 Celularity, Inc. Tumor suppression using human placenta-derived intermediate natural killer (pink) cells in combination with an antibody
MX2020003314A (en) 2017-10-02 2020-10-28 Gamida Cell Ltd Expansion and use of expanded nk cell fractions.
CN108220237B (en) * 2017-12-20 2020-11-06 中国科学院遗传与发育生物学研究所 Human NK/T cell line
NZ766453A (en) 2018-02-01 2022-02-25 Nkmax Co Ltd Method of producing natural killer cells and composition for treating cancer
CN108486054B (en) * 2018-04-12 2022-04-19 平安爱普德(北京)生物技术有限公司 Method for in vitro amplification of I-type natural cytotoxic lymphocyte ILC1/NK
EP3790562A4 (en) * 2018-05-11 2022-01-12 The Regents of the University of California Modification of immune cells to increase activity
WO2019231243A1 (en) * 2018-05-29 2019-12-05 사회복지법인 삼성생명공익재단 Feeder cell expressing ox40l and method for culturing natural killer cells using same
US20210267190A1 (en) * 2018-07-10 2021-09-02 Nantkwest, Inc. Cryopreservation
CN109294985B (en) 2018-10-25 2022-02-18 江苏普瑞康生物医药科技有限公司 Culture medium system for NK cell in-vitro amplification and NK cell in-vitro amplification method
CN109337870B (en) * 2018-12-24 2020-07-03 广东暨德康民生物科技有限责任公司 Human V gamma 9V delta 2T cell amplification method and culture medium
EP3773918A4 (en) 2019-03-05 2022-01-05 Nkarta, Inc. Cd19-directed chimeric antigen receptors and uses thereof in immunotherapy
CA3136348A1 (en) 2019-04-09 2020-10-15 Nurix Therapeutics, Inc. 3-substituted piperidine compounds for cbl-b inhibition, and use of a cbl-b inhibitor in combination with a cancer vaccine and/or oncolytic virus
EP3969447A1 (en) 2019-05-17 2022-03-23 Nurix Therapeutics, Inc. Cyano cyclobutyl compounds for cbl-b inhibition and uses thereof
WO2020252441A2 (en) * 2019-06-14 2020-12-17 Gumrukcu Serhat Activated lymphocytic cells and methods of using the same to treat cancer and infectious conditions
KR20220026581A (en) 2019-06-26 2022-03-04 누릭스 테라퓨틱스 인코포레이티드 Substituted benzyl-triazole compounds for CBL-B inhibition, and further uses thereof
US11453862B2 (en) 2019-07-08 2022-09-27 Immunitybio, Inc. Mononuclear cell derived NK cells
US20220249567A1 (en) * 2019-07-22 2022-08-11 Glycostem Therapeutics B.V. Low density cell culture
WO2021021761A1 (en) 2019-07-30 2021-02-04 Nurix Therapeutics, Inc. Urea, amide, and substituted heteroaryl compounds for cbl-b inhibition
JP2021136883A (en) * 2020-03-02 2021-09-16 株式会社ガイアバイオメディシン Method for treating highly active nk cells
US11547726B2 (en) * 2020-03-26 2023-01-10 Honed Life Sciences, Llc Enhancement of production of NK cells from stem cells
CN111500535B (en) * 2020-04-30 2023-04-14 惠和生物技术(上海)有限公司 Method and culture medium for in vitro culture of human natural killer cells
CN112251405A (en) * 2020-10-15 2021-01-22 大连天星生物技术有限责任公司 Method for efficiently inducing and amplifying NK cells in vitro
AU2021374598A1 (en) * 2020-11-03 2023-06-22 Edigene (Guangzhou) Inc. Cell preparation, use of protein in characterizing hematopoietic stem cells, and method for determining hematopoietic stem cells
US11459372B2 (en) 2020-11-30 2022-10-04 Crispr Therapeutics Ag Gene-edited natural killer cells
TW202241935A (en) 2020-12-18 2022-11-01 美商世紀治療股份有限公司 Chimeric antigen receptor system with adaptable receptor specificity
EP4271798A1 (en) 2020-12-30 2023-11-08 CRISPR Therapeutics AG Compositions and methods for differentiating stem cells into nk cells
AU2022219942A1 (en) * 2021-02-09 2023-08-17 Takeda Pharmaceutical Company Limited Methods and compositions for cryopreservation of immune cells
IL305012A (en) * 2021-02-09 2023-10-01 Takeda Pharmaceuticals Co Methods and compositions for freezing and thawing mammalian cells
WO2022173737A1 (en) * 2021-02-09 2022-08-18 Millennium Pharmaceuticals, Inc. Methods and compositions for freezing and thawing mammalian cells
AR124835A1 (en) * 2021-02-09 2023-05-10 Millennium Pharm Inc METHODS AND COMPOSITIONS FOR THE CRYOPRESERVATION OF IMMUNE CELLS
WO2022226337A1 (en) * 2021-04-22 2022-10-27 Artec Biotech, Inc. Method for producing hematopoietic cells from stem cells using vascular organoids
CN115590013A (en) * 2021-07-07 2023-01-13 北京鼎成肽源生物技术有限公司(Cn) Cryopreservation solution and cryopreservation method for natural killer cells and application of cryopreservation solution and cryopreservation method
CN113416701B (en) * 2021-07-28 2023-05-09 新疆西部赛澳生物科技有限责任公司 NK cell culture medium and culture method
CN115261318A (en) * 2021-09-29 2022-11-01 苏州艾凯利元生物科技有限公司 Method for producing natural killer cells
WO2023081813A1 (en) 2021-11-05 2023-05-11 St. Jude Children's Research Hospital, Inc. Zip cytokine receptors
CN114807031A (en) * 2022-05-13 2022-07-29 山东赛恩福干细胞工程集团有限公司 Construction method of human peripheral blood immune cell bank and stem cell bank
WO2023240182A1 (en) 2022-06-08 2023-12-14 St. Jude Children's Research Hospital, Inc. Disruption of kdm4a in t cells to enhance immunotherapy
CN114736859B (en) * 2022-06-13 2022-08-19 广东先康达生物科技有限公司 Culture solution and culture method for cord blood NK cells
CN115094037B (en) * 2022-07-19 2023-11-10 百欧派(天津)生物技术有限公司 Composition with canine natural killer cell in-vitro activation function matched with autologous plasma for use and application thereof
WO2024059787A1 (en) 2022-09-16 2024-03-21 St. Jude Children's Research Hospital, Inc. Disruption of asxl1 in t cells to enhance immunotherapy

Family Cites Families (291)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3862002A (en) 1962-05-08 1975-01-21 Sanfar Lab Inc Production of physiologically active placental substances
US4829000A (en) 1985-08-30 1989-05-09 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Reconstituted basement membrane complex with biological activity
US4798824A (en) 1985-10-03 1989-01-17 Wisconsin Alumni Research Foundation Perfusate for the preservation of organs
US5266480A (en) 1986-04-18 1993-11-30 Advanced Tissue Sciences, Inc. Three-dimensional skin culture system
US5902741A (en) 1986-04-18 1999-05-11 Advanced Tissue Sciences, Inc. Three-dimensional cartilage cultures
US5863531A (en) 1986-04-18 1999-01-26 Advanced Tissue Sciences, Inc. In vitro preparation of tubular tissue structures by stromal cell culture on a three-dimensional framework
NZ226750A (en) 1987-10-29 1990-09-26 Amrad Corp Ltd Immortalisation of neural precursor cells by introducing a retrovirus vector containing a myc-oncogene
US5192553A (en) 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US5004681B1 (en) 1987-11-12 2000-04-11 Biocyte Corp Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
GB8803697D0 (en) 1988-02-17 1988-03-16 Deltanine Research Ltd Clinical developments using amniotic membrane cells
US5284766A (en) 1989-02-10 1994-02-08 Kao Corporation Bed material for cell culture
FR2646438B1 (en) 1989-03-20 2007-11-02 Pasteur Institut A METHOD FOR SPECIFIC REPLACEMENT OF A COPY OF A GENE PRESENT IN THE RECEIVER GENOME BY INTEGRATION OF A GENE DIFFERENT FROM THAT OR INTEGRATION
US5399493A (en) 1989-06-15 1995-03-21 The Regents Of The University Of Michigan Methods and compositions for the optimization of human hematopoietic progenitor cell cultures
US5605822A (en) 1989-06-15 1997-02-25 The Regents Of The University Of Michigan Methods, compositions and devices for growing human hematopoietic cells
US5635386A (en) 1989-06-15 1997-06-03 The Regents Of The University Of Michigan Methods for regulating the specific lineages of cells produced in a human hematopoietic cell culture
US5763266A (en) 1989-06-15 1998-06-09 The Regents Of The University Of Michigan Methods, compositions and devices for maintaining and growing human stem and/or hematopoietics cells
US5437994A (en) 1989-06-15 1995-08-01 Regents Of The University Of Michigan Method for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
DE69034263D1 (en) 1989-07-25 2009-04-02 Cell Genesys Inc Homologous recombination for universal donor cells and mammalian chimeric cells
US5464764A (en) 1989-08-22 1995-11-07 University Of Utah Research Foundation Positive-negative selection methods and vectors
DK0747485T3 (en) 1989-11-06 1999-08-16 Cell Genesys Inc Preparation of proteins using homologous recombination
US5272071A (en) 1989-12-22 1993-12-21 Applied Research Systems Ars Holding N.V. Method for the modification of the expression characteristics of an endogenous gene of a given cell line
US5061620A (en) 1990-03-30 1991-10-29 Systemix, Inc. Human hematopoietic stem cell
US5635387A (en) 1990-04-23 1997-06-03 Cellpro, Inc. Methods and device for culturing human hematopoietic cells and their precursors
US6326198B1 (en) 1990-06-14 2001-12-04 Regents Of The University Of Michigan Methods and compositions for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US5733542A (en) 1990-11-16 1998-03-31 Haynesworth; Stephen E. Enhancing bone marrow engraftment using MSCS
US5197985A (en) 1990-11-16 1993-03-30 Caplan Arnold I Method for enhancing the implantation and differentiation of marrow-derived mesenchymal cells
US5811094A (en) 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US5226914A (en) 1990-11-16 1993-07-13 Caplan Arnold I Method for treating connective tissue disorders
US5837539A (en) 1990-11-16 1998-11-17 Osiris Therapeutics, Inc. Monoclonal antibodies for human mesenchymal stem cells
US5486359A (en) 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US6010696A (en) 1990-11-16 2000-01-04 Osiris Therapeutics, Inc. Enhancing hematopoietic progenitor cell engraftment using mesenchymal stem cells
US5190556A (en) 1991-03-19 1993-03-02 O.B. Tech, Inc. Cord cutter sampler
US5744361A (en) 1991-04-09 1998-04-28 Indiana University Expansion of human hematopoietic progenitor cells in a liquid medium
EP0552380A4 (en) 1991-08-08 1995-01-25 Kao Corp Cell culture support, production thereof, and production of cell cluster using same
EP0529751A1 (en) 1991-08-09 1993-03-03 W.R. Grace & Co.-Conn. Cell culture substrate, test material for cell culture and preparations thereof
WO1993004169A1 (en) 1991-08-20 1993-03-04 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
US5356373A (en) 1991-11-15 1994-10-18 Miles Inc. Method and apparatus for autologous transfusions in premature infants
US6057123A (en) 1991-12-23 2000-05-02 British Biotech Pharmaceuticals Limited Stem cell inhibiting proteins
US5668104A (en) 1992-03-31 1997-09-16 Toray Industries, Inc. Hematopoietic stem cell growth-promoting compositions containing a fibroblast-derived fragment of fibronectin and a growth factor, and methods employing them in vitro or in vivo
US5552267A (en) 1992-04-03 1996-09-03 The Trustees Of Columbia University In The City Of New York Solution for prolonged organ preservation
US5436151A (en) 1992-04-03 1995-07-25 Regents Of The University Of Minnesota Method for culturing human hematopoietic stem cells in vitro
US5460964A (en) 1992-04-03 1995-10-24 Regents Of The University Of Minnesota Method for culturing hematopoietic cells
AU4543193A (en) 1992-06-22 1994-01-24 Henry E. Young Scar inhibitory factor and use thereof
US5672346A (en) 1992-07-27 1997-09-30 Indiana University Foundation Human stem cell compositions and methods
US5693482A (en) 1992-07-27 1997-12-02 California Institute Of Technology Neural chest stem cell assay
US5849553A (en) 1992-07-27 1998-12-15 California Institute Of Technology Mammalian multipotent neural stem cells
EP0669974A1 (en) 1992-11-16 1995-09-06 Rhone-Poulenc Rorer Pharmaceuticals Inc. Pluripotential quiescent stem cell population
US5772992A (en) 1992-11-24 1998-06-30 G.D. Searle & Co. Compositions for co-administration of interleukin-3 mutants and other cytokines and hematopoietic factors
US5654186A (en) 1993-02-26 1997-08-05 The Picower Institute For Medical Research Blood-borne mesenchymal cells
JPH08510997A (en) 1993-03-31 1996-11-19 プロ−ニューロン,インコーポレイテッド Stem cell growth inhibitors and uses thereof
GB9308271D0 (en) 1993-04-21 1993-06-02 Univ Edinburgh Method of isolating and/or enriching and/or selectively propagating pluripotential animal cells and animals for use in said method
US5709854A (en) 1993-04-30 1998-01-20 Massachusetts Institute Of Technology Tissue formation by injecting a cell-polymeric solution that gels in vivo
US5372581A (en) 1993-07-21 1994-12-13 Minneapolis Children's Services Corporation Method and apparatus for placental blood collection
IL107483A0 (en) 1993-11-03 1994-02-27 Yeda Res & Dev Bone marrow transplantation
US5591625A (en) 1993-11-24 1997-01-07 Case Western Reserve University Transduced mesenchymal stem cells
US6288030B1 (en) 1993-12-22 2001-09-11 Amgen Inc. Stem cell factor formulations and methods
US6001654A (en) 1994-01-28 1999-12-14 California Institute Of Technology Methods for differentiating neural stem cells to neurons or smooth muscle cells using TGT-β super family growth factors
US5942496A (en) 1994-02-18 1999-08-24 The Regent Of The University Of Michigan Methods and compositions for multiple gene transfer into bone cells
US6174333B1 (en) 1994-06-06 2001-01-16 Osiris Therapeutics, Inc. Biomatrix for soft tissue regeneration using mesenchymal stem cells
DE69531638T2 (en) 1994-06-06 2004-06-17 Osiris Therapeutics, Inc. BIOMATRIX FOR TISSUE REGENATION
DE4422667A1 (en) 1994-06-30 1996-01-04 Boehringer Ingelheim Int Process for the production and cultivation of hematopoietic progenitor cells
US6103522A (en) 1994-07-20 2000-08-15 Fred Hutchinson Cancer Research Center Human marrow stromal cell lines which sustain hematopoiesis
US5516532A (en) 1994-08-05 1996-05-14 Children's Medical Center Corporation Injectable non-immunogenic cartilage and bone preparation
US5827742A (en) 1994-09-01 1998-10-27 Beth Israel Deaconess Medical Center, Inc. Method of selecting pluripotent hematopioetic progenitor cells
US5665557A (en) 1994-11-14 1997-09-09 Systemix, Inc. Method of purifying a population of cells enriched for hematopoietic stem cells populations of cells obtained thereby and methods of use thereof
MX9701512A (en) 1994-11-16 1997-05-31 Amgen Inc Use of stem cell factor and soluble interleukin-6 receptor for the ex vivo expansion of hematopoietic multipotential cells.
US5874301A (en) 1994-11-21 1999-02-23 National Jewish Center For Immunology And Respiratory Medicine Embryonic cell populations and methods to isolate such populations
US5914268A (en) 1994-11-21 1999-06-22 National Jewish Center For Immunology & Respiratory Medicine Embryonic cell populations and methods to isolate such populations
US5789147A (en) 1994-12-05 1998-08-04 New York Blood Center, Inc. Method for concentrating white cells from whole blood by adding a red cell sedimentation reagent to whole anticoagulated blood
US5736396A (en) 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US5695998A (en) 1995-02-10 1997-12-09 Purdue Research Foundation Submucosa as a growth substrate for islet cells
US6011000A (en) 1995-03-03 2000-01-04 Perrine; Susan P. Compositions for the treatment of blood disorders
US5906934A (en) 1995-03-14 1999-05-25 Morphogen Pharmaceuticals, Inc. Mesenchymal stem cells for cartilage repair
US5716616A (en) 1995-03-28 1998-02-10 Thomas Jefferson University Isolated stromal cells for treating diseases, disorders or conditions characterized by bone defects
US5677139A (en) 1995-04-21 1997-10-14 President And Fellows Of Harvard College In vitro differentiation of CD34+ progenitor cells into T lymphocytes
US5733541A (en) 1995-04-21 1998-03-31 The Regent Of The University Of Michigan Hematopoietic cells: compositions and methods
HUP9801443A3 (en) 1995-04-27 2001-11-28 Procter & Gamble Carrier substrate treated with high internal water phase inverse emulsion made with an organopolysiloxane-polyoxyalkylene emulsifier
US5925567A (en) 1995-05-19 1999-07-20 T. Breeders, Inc. Selective expansion of target cell populations
US5908782A (en) 1995-06-05 1999-06-01 Osiris Therapeutics, Inc. Chemically defined medium for human mesenchymal stem cells
US5830708A (en) 1995-06-06 1998-11-03 Advanced Tissue Sciences, Inc. Methods for production of a naturally secreted extracellular matrix
WO1996040875A1 (en) 1995-06-07 1996-12-19 Novartis Ag Methods for obtaining compositions enriched for hematopoietic stem cells and antibodies for use therein
US5654381A (en) 1995-06-16 1997-08-05 Massachusetts Institute Of Technology Functionalized polyester graft copolymers
US5877299A (en) 1995-06-16 1999-03-02 Stemcell Technologies Inc. Methods for preparing enriched human hematopoietic cell preparations
US6306575B1 (en) 1995-06-16 2001-10-23 Stemcell Technologies, Inc. Methods for preparing enriched human hematopoietic cell preparations
US5858782A (en) 1995-11-13 1999-01-12 Regents Of The University Of Michigan Functional human hematopoietic cells
US5922597A (en) 1995-11-14 1999-07-13 Regents Of The University Of Minnesota Ex vivo culture of stem cells
PT868505E (en) 1995-11-16 2005-06-30 Univ Case Western Reserve IN VITRO CONDROGENIC INDUCTION OF HUMAN MESENCHINEAL STAMINA CELLS
US6337387B1 (en) 1995-11-17 2002-01-08 Asahi Kasei Kabushiki Kaisha Differentiation-suppressive polypeptide
US5716794A (en) 1996-03-29 1998-02-10 Xybernaut Corporation Celiac antigen
AU731468B2 (en) 1996-04-19 2001-03-29 Mesoblast International Sarl Regeneration and augmentation of bone using mesenchymal stem cells
JP2000508922A (en) 1996-04-26 2000-07-18 ケース ウエスターン リザーブ ユニバーシティ Skin regeneration using mesenchymal stem cells
US5919176A (en) 1996-05-14 1999-07-06 Children's Hospital Medical Center Of Northern California Apparatus and method for collecting blood from an umbilical cord
US5827740A (en) 1996-07-30 1998-10-27 Osiris Therapeutics, Inc. Adipogenic differentiation of human mesenchymal stem cells
US5851984A (en) 1996-08-16 1998-12-22 Genentech, Inc. Method of enhancing proliferation or differentiation of hematopoietic stem cells using Wnt polypeptides
US5916202A (en) 1996-08-30 1999-06-29 Haswell; John N. Umbilical cord blood collection
US6227202B1 (en) 1996-09-03 2001-05-08 Maulana Azad Medical College Method of organogenesis and tissue regeneration/repair using surgical techniques
US5945337A (en) 1996-10-18 1999-08-31 Quality Biological, Inc. Method for culturing CD34+ cells in a serum-free medium
US5919702A (en) 1996-10-23 1999-07-06 Advanced Tissue Science, Inc. Production of cartilage tissue using cells isolated from Wharton's jelly
US5969105A (en) 1996-10-25 1999-10-19 Feng; Yiqing Stem cell factor receptor agonists
US6335195B1 (en) 1997-01-28 2002-01-01 Maret Corporation Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US6152142A (en) 1997-02-28 2000-11-28 Tseng; Scheffer C. G. Grafts made from amniotic membrane; methods of separating, preserving, and using such grafts in surgeries
US6231880B1 (en) 1997-05-30 2001-05-15 Susan P. Perrine Compositions and administration of compositions for the treatment of blood disorders
US6261549B1 (en) 1997-07-03 2001-07-17 Osiris Therapeutics, Inc. Human mesenchymal stem cells from peripheral blood
US7514074B2 (en) 1997-07-14 2009-04-07 Osiris Therapeutics, Inc. Cardiac muscle regeneration using mesenchymal stem cells
CA2296704C (en) 1997-07-14 2010-10-19 Osiris Therapeutics, Inc. Cardiac muscle regeneration using mesenchymal stem cells
US6077708A (en) 1997-07-18 2000-06-20 Collins; Paul C. Method of determining progenitor cell content of a hematopoietic cell culture
US5879318A (en) 1997-08-18 1999-03-09 Npbi International B.V. Method of and closed system for collecting and processing umbilical cord blood
WO1999011287A1 (en) 1997-09-04 1999-03-11 Osiris Therapeutics, Inc. Ligands that modulate differentiation of mesenchymal stem cells
US5968829A (en) 1997-09-05 1999-10-19 Cytotherapeutics, Inc. Human CNS neural stem cells
US6093531A (en) 1997-09-29 2000-07-25 Neurospheres Holdings Ltd. Generation of hematopoietic cells from multipotent neural stem cells
US6248587B1 (en) 1997-11-26 2001-06-19 University Of Southern Cailfornia Method for promoting mesenchymal stem and lineage-specific cell proliferation
US6059968A (en) 1998-01-20 2000-05-09 Baxter International Inc. Systems for processing and storing placenta/umbilical cord blood
US6291240B1 (en) 1998-01-29 2001-09-18 Advanced Tissue Sciences, Inc. Cells or tissues with increased protein factors and methods of making and using same
CA2323073C (en) 1998-03-13 2010-06-22 Osiris Therapeutics, Inc. Uses for human non-autologous mesenchymal stem cells
ATE316795T1 (en) 1998-03-18 2006-02-15 Osiris Therapeutics Inc MESENCHYMAL STEM CELLS FOR THE PREVENTION AND TREATMENT OF IMMUNE RESPONSE DURING TRANSPLANTATIONS
US6368636B1 (en) 1998-03-18 2002-04-09 Osiris Therapeutics, Inc. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
DE69910362T2 (en) 1998-04-03 2004-06-24 Osiris Therapeutics, Inc. APPLICATION OF THE MESENCHYMAL STEM CELLS AS IMMUNE SUPPRESSIVA
JP2002513762A (en) 1998-05-04 2002-05-14 ポイント セラピューティクス, インコーポレイテッド Hematopoietic stimulation
US6835377B2 (en) 1998-05-13 2004-12-28 Osiris Therapeutics, Inc. Osteoarthritis cartilage regeneration
US6225119B1 (en) 1998-05-22 2001-05-01 Osiris Therapeutics, Inc. Production of megakaryocytes by the use of human mesenchymal stem cells
US6387367B1 (en) 1998-05-29 2002-05-14 Osiris Therapeutics, Inc. Human mesenchymal stem cells
AU4336599A (en) 1998-06-08 1999-12-30 Osiris Therapeutics, Inc. (in vitro) maintenance of hematopoietic stem cells
US6255112B1 (en) 1998-06-08 2001-07-03 Osiris Therapeutics, Inc. Regulation of hematopoietic stem cell differentiation by the use of human mesenchymal stem cells
US6713245B2 (en) 1998-07-06 2004-03-30 Diacrin, Inc. Methods for storing neural cells such that they are suitable for transplantation
WO2000006150A1 (en) 1998-07-28 2000-02-10 Synergen Ag Use of creatine compounds for treatment of bone or cartilage cells and tissues
US5958767A (en) 1998-08-14 1999-09-28 The Children's Medical Center Corp. Engraftable human neural stem cells
JP3517359B2 (en) 1998-09-14 2004-04-12 テルモ株式会社 Cell separation / collection apparatus and cell separation / collection method
WO2000017325A1 (en) 1998-09-23 2000-03-30 Mount Sinai Hospital Trophoblast cell preparations
WO2000027995A1 (en) 1998-11-09 2000-05-18 Monash University Embryonic stem cells
US6548299B1 (en) 1999-11-12 2003-04-15 Mark J. Pykett Lymphoid tissue-specific cell production from hematopoietic progenitor cells in three-dimensional devices
AU1720400A (en) 1998-11-12 2000-05-29 Cell Science Therapeutics, Inc. Lymphoid tissue-specific cell production from hematopoietic progenitor cells in three-dimensional devices
US6184035B1 (en) 1998-11-18 2001-02-06 California Institute Of Technology Methods for isolation and activation of, and control of differentiation from, skeletal muscle stem or progenitor cells
US6102871A (en) 1998-11-23 2000-08-15 Coe; Rosemarie O. Blood collection funnel
US6328765B1 (en) 1998-12-03 2001-12-11 Gore Enterprise Holdings, Inc. Methods and articles for regenerating living tissue
ES2224726T3 (en) 1998-12-24 2005-03-01 Biosafe S.A. BLOOD SEPARATION SYSTEM INDICATED IN PARTICULAR FOR THE CONCENTRATION OF HEMATOPOYETIC MOTHER CELLS.
US20030007954A1 (en) 1999-04-12 2003-01-09 Gail K. Naughton Methods for using a three-dimensional stromal tissue to promote angiogenesis
IN191359B (en) 1999-04-20 2003-11-29 Nat Inst Immunology
AU4860900A (en) 1999-06-02 2000-12-18 Lifebank Services, L.L.C. Methods of isolation, cryopreservation, and therapeutic use of human amniotic epithelial cells
US6333029B1 (en) 1999-06-30 2001-12-25 Ethicon, Inc. Porous tissue scaffoldings for the repair of regeneration of tissue
US7015037B1 (en) 1999-08-05 2006-03-21 Regents Of The University Of Minnesota Multiponent adult stem cells and methods for isolation
US8147824B2 (en) 1999-08-05 2012-04-03 Athersys, Inc. Immunomodulatory properties of multipotent adult progenitor cells and uses thereof
US8075881B2 (en) 1999-08-05 2011-12-13 Regents Of The University Of Minnesota Use of multipotent adult stem cells in treatment of myocardial infarction and congestive heart failure
US6239157B1 (en) 1999-09-10 2001-05-29 Osiris Therapeutics, Inc. Inhibition of osteoclastogenesis
US6685936B2 (en) 1999-10-12 2004-02-03 Osiris Therapeutics, Inc. Suppressor cells induced by culture with mesenchymal stem cells for treatment of immune responses in transplantation
US6280718B1 (en) 1999-11-08 2001-08-28 Wisconsin Alumni Reasearch Foundation Hematopoietic differentiation of human pluripotent embryonic stem cells
DE19954421A1 (en) 1999-11-12 2001-05-31 Lohmann Therapie Syst Lts Film-like preparation for the biphase release of pharmacologically active or other substances
EP1264877B1 (en) 2000-03-16 2013-10-02 Cellseed Inc. Cell cultivation-support material, method of cocultivation of cells and cocultivated cell sheet obtained therefrom
WO2001075094A1 (en) 2000-04-04 2001-10-11 Thomas Jefferson University Application of myeloid-origin cells to the nervous system
EP1289557B1 (en) 2000-06-06 2006-07-12 Glaxo Group Limited Cancer treatment composition containing an anti-neoplastic agent and a pde4 inhibitor
US20050009876A1 (en) 2000-07-31 2005-01-13 Bhagwat Shripad S. Indazole compounds, compositions thereof and methods of treatment therewith
US20080152629A1 (en) 2000-12-06 2008-06-26 James Edinger Placental stem cell populations
KR100915482B1 (en) 2000-12-06 2009-09-03 하리리 로버트 제이 Method of collecting placental stem cells
US7311905B2 (en) 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
US20030045552A1 (en) 2000-12-27 2003-03-06 Robarge Michael J. Isoindole-imide compounds, compositions, and uses thereof
US7091353B2 (en) 2000-12-27 2006-08-15 Celgene Corporation Isoindole-imide compounds, compositions, and uses thereof
EP1362095B1 (en) 2001-02-14 2015-05-27 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
NZ528035A (en) 2001-02-14 2005-07-29 Robert J Hariri Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
EP1367899A4 (en) 2001-02-14 2004-07-28 Leo T Furcht Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
US20020132343A1 (en) 2001-03-19 2002-09-19 Clark Lum System and method for delivering umbilical cord-derived tissue-matched stem cells for transplantation
US20030044977A1 (en) 2001-08-10 2003-03-06 Norio Sakuragawa Human stem cells originated from human amniotic mesenchymal cell layer
DE10139783C1 (en) 2001-08-14 2003-04-17 Transtissue Technologies Gmbh Cell compositions for the treatment of osteoarthritis, and methods for their production
WO2003018780A1 (en) 2001-08-27 2003-03-06 Advanced Cell Technology, Inc. De-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
EP1288293A1 (en) 2001-08-30 2003-03-05 Norio Sakuragawa Human neural stem cells originated from human amniotic mesenchymal cell layer
CN1195055C (en) 2001-09-06 2005-03-30 周胜利 Method for establishing hematopoietic stem cells bank by extracting hematopoietic cells from placenta tissues
US20030068306A1 (en) 2001-09-14 2003-04-10 Dilber Mehmet Sirac Medium
US9969980B2 (en) 2001-09-21 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
DE60233248D1 (en) 2001-11-15 2009-09-17 Childrens Medical Center PROCESS FOR THE ISOLATION, EXPANSION AND DIFFERENTIATION OF FEDERAL STRAIN CELLS FROM CHORION ZOTTE, FRUIT WATER AND PLAZENTA AND THERAPEUTIC USES THEREOF
JP3728750B2 (en) 2001-11-22 2005-12-21 ニプロ株式会社 Cultured skin and method for producing the same
US7799324B2 (en) 2001-12-07 2010-09-21 Geron Corporation Using undifferentiated embryonic stem cells to control the immune system
JP3934539B2 (en) 2001-12-12 2007-06-20 独立行政法人科学技術振興機構 Adult or postnatal tissue progenitor cells derived from placenta
WO2003061591A2 (en) 2002-01-22 2003-07-31 Advanced Cell Technology, Inc. Stem cell-derived endothelial cells modified to disrupt tumor angiogenesis
MXPA04007732A (en) 2002-02-13 2004-10-15 Anthrogenesis Corp Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells.
US20030161818A1 (en) 2002-02-25 2003-08-28 Kansas State University Research Foundation Cultures, products and methods using stem cells
US7736892B2 (en) 2002-02-25 2010-06-15 Kansas State University Research Foundation Cultures, products and methods using umbilical cord matrix cells
WO2003077865A2 (en) 2002-03-15 2003-09-25 Baxter International Inc. Methods and compositions for directing cells to target organs
US20030187515A1 (en) 2002-03-26 2003-10-02 Hariri Robert J. Collagen biofabric and methods of preparing and using the collagen biofabric
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
KR20050000398A (en) 2002-04-12 2005-01-03 셀진 코포레이션 Methods for identification of modulators of angiogenesis, compounds discovered thereby, and methods of treatment using the compounds
CA2481385A1 (en) 2002-04-12 2003-10-23 Celgene Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US20040161419A1 (en) 2002-04-19 2004-08-19 Strom Stephen C. Placental stem cells and uses thereof
CA2483010A1 (en) 2002-04-19 2003-10-30 University Of Pittsburgh Of The Commonwealth System Of Higher Education Placental derived stem cells and uses thereof
EP1525308A4 (en) 2002-05-30 2006-11-02 Celgene Corp Methods of using jnk or mkk inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes
US7422736B2 (en) 2002-07-26 2008-09-09 Food Industry Research And Development Institute Somatic pluripotent cells
JP4603883B2 (en) 2002-07-31 2010-12-22 サントル・ナショナル・ドゥ・ラ・レシェルシュ・サイエンティフィーク−セ・エン・エール・エス− Stem cells derived from adipose tissue and cells differentiated from the cells
EP1535994A4 (en) 2002-08-23 2005-12-07 Srl Inc Human bone stem cells
US20040062753A1 (en) 2002-09-27 2004-04-01 Alireza Rezania Composite scaffolds seeded with mammalian cells
EP1571910A4 (en) 2002-11-26 2009-10-28 Anthrogenesis Corp Cytotherapeutics, cytotherapeutic units and methods for treatments using them
DE602004028803D1 (en) 2003-02-11 2010-10-07 John E Davies PRECURSOR CELLS FROM THE WHARTON SULCE OF HUMAN NECK LOCKS
NZ542127A (en) 2003-02-13 2008-04-30 Anthrogenesis Corp Use of umbilical cord blood to treat individuals having a disease, disorder or condition
WO2004087896A2 (en) 2003-03-31 2004-10-14 Pfizer Products Inc. Hepatocyte differentiation of stem cells
CN1548529A (en) 2003-05-09 2004-11-24 中国人民解放军军事医学科学院基础医 Separation method of buffering stem cell in human placenta
WO2005038012A2 (en) 2003-06-27 2005-04-28 Ethicon Incorporated Cartilage and bone repair and regeneration using postpartum-derived cells
US7875272B2 (en) 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US7569385B2 (en) 2003-08-14 2009-08-04 The Regents Of The University Of California Multipotent amniotic fetal stem cells
US20050089513A1 (en) 2003-10-28 2005-04-28 Norio Sakuragawa Side population cells originated from human amnion and their uses
WO2005047491A2 (en) 2003-11-10 2005-05-26 Amgen Inc. Methods of using g-csf mobilized c-kit+cells in the production of embryoid body-like cell clusters for tissue repair and in the treatment of cardiac myopathy
KR100560340B1 (en) 2003-11-11 2006-03-14 한훈 Method of isolating and culturing mesenchymal stem cell derived from umbilical cord blood
JP2005151907A (en) 2003-11-27 2005-06-16 Shigeo Saito Human stem cell derived from placenta or amnion and method for establishing the same and method for differentiation-induction to organ
TWI338714B (en) 2003-12-02 2011-03-11 Cathay General Hospital Method of isolation and enrichment of mesenchymal stem cells from amniotic fluid
KR20110116225A (en) 2003-12-02 2011-10-25 셀진 코포레이션 Methods and compositions for the treatment and management of hemoglobinopathy and anemia
US20050176139A1 (en) 2004-01-12 2005-08-11 Yao-Chang Chen Placental stem cell and methods thereof
US20050266391A1 (en) 2004-01-15 2005-12-01 Bennett Brydon L Methods for preserving tissue
WO2005093044A1 (en) 2004-03-22 2005-10-06 Osiris Therapeutics, Inc. Mesenchymal stem cells and uses therefor
NZ550027A (en) 2004-03-26 2009-03-31 Celgene Corp Systems and methods for providing a stem cell bank
WO2005105982A1 (en) 2004-03-31 2005-11-10 Reinhardt Schwartz-Albiez Method for the ex vivo sequential expansion of human postembryonic stem cells and subsequent selective differentiation
WO2005105992A1 (en) 2004-04-21 2005-11-10 New York Eye & Ear Infirmary Chondrocyte culture formulations
US20090297471A1 (en) * 2004-05-28 2009-12-03 Mayo Foundation For Medical Education And Research Methods For Autologous Stem Cell Transplantation
JP2006006249A (en) 2004-06-28 2006-01-12 Hiroshima Univ Method for culturing amnion-derived cell and utilization of the same
US7244759B2 (en) 2004-07-28 2007-07-17 Celgene Corporation Isoindoline compounds and methods of making and using the same
US20080292597A1 (en) 2004-07-29 2008-11-27 David A Steenblock Umbilical Cord Stem Cell Composition & Method of Treating Neurological Diseases
CA2971062C (en) 2004-08-16 2019-02-26 Cellresearch Corporation Pte Ltd Isolation of stem/progenitor cells from amniotic membrane of umbilical cord
US7909806B2 (en) 2004-09-23 2011-03-22 Anthrogenesis Corporation Cord blood and placenta collection kit
US7147626B2 (en) 2004-09-23 2006-12-12 Celgene Corporation Cord blood and placenta collection kit
US8039258B2 (en) 2004-09-28 2011-10-18 Ethicon, Inc. Tissue-engineering scaffolds containing self-assembled-peptide hydrogels
US20060166361A1 (en) 2004-12-21 2006-07-27 Agnieszka Seyda Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
US20060171930A1 (en) 2004-12-21 2006-08-03 Agnieszka Seyda Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
CA2589063C (en) 2004-12-23 2016-08-09 Ethicon Incorporated Treatment of parkinson's disease and related disorders using postpartum derived cells
PL1831356T3 (en) 2004-12-23 2017-07-31 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US9056093B2 (en) 2005-01-07 2015-06-16 Wake Forest University Health Sciences Regeneration of pancreatic islets by amniotic fluid stem cell therapy
WO2006091766A2 (en) 2005-02-24 2006-08-31 Jau-Nan Lee Human trophoblast stem cells and use thereof
US20060222634A1 (en) 2005-03-31 2006-10-05 Clarke Diana L Amnion-derived cell compositions, methods of making and uses thereof
WO2006111706A1 (en) 2005-04-16 2006-10-26 Axordia Limited Cytotrophoblast stem cell
AU2006202209B2 (en) 2005-05-27 2011-04-14 Lifescan, Inc. Amniotic fluid derived cells
ZA200800144B (en) 2005-06-10 2010-02-24 Celgene Corp Human placental collagen compositions, processes for their preparation, methods of their use and kits comprising the compositions
KR20080026198A (en) 2005-06-30 2008-03-24 안트로제네시스 코포레이션 Repair of tympanic membrane using placenta derived collagen biofabric
WO2007009062A2 (en) 2005-07-13 2007-01-18 Anthrogenesis Corporation Treatment of leg ulcers using placenta derived collagen biofabric
WO2007009061A2 (en) 2005-07-13 2007-01-18 Anthrogenesis Corporation Ocular plug formed from placenta derived collagen biofabric
WO2007011693A2 (en) 2005-07-14 2007-01-25 Medistem Laboratories, Inc. Compositions of placentally-derived stem cells for the treatment of cancer
US20080226612A1 (en) 2005-08-19 2008-09-18 Bio Regenerate, Inc. Compositions of Cells Enriched for Combinations of Various Stem and Progenitor Cell Populations, Methods of Use Thereof and Methods of Private Banking Thereof
CN101258160A (en) * 2005-09-12 2008-09-03 全南大学校产学协力团 Natural killer cell compositions and method for production of the same
WO2007032634A1 (en) * 2005-09-12 2007-03-22 Industry Foundation Of Chonnam National University A method for production of mature natural killer cell
ES2502841T3 (en) * 2005-09-28 2014-10-06 Ipd-Therapeutics B.V. Methods and means for stem cell proliferation and subsequent generation and expansion of progenitor cells, as well as production of effector cells as clinical therapeutics
ES2452595T3 (en) 2005-10-13 2014-04-02 Anthrogenesis Corporation Immunomodulation using placental stem cells
EP2368973A1 (en) 2005-10-13 2011-09-28 Anthrogenesis Corporation Production Of Oligodendrocytes From Placenta-Derived Stem Cells
CN100344757C (en) 2005-10-18 2007-10-24 天津昂赛细胞基因工程有限公司 Human placenta, umbilical cord mesenchyma stemcell stock and its construction method
WO2007070870A1 (en) 2005-12-16 2007-06-21 Ethicon, Inc. Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation
EP1976975B1 (en) 2005-12-19 2012-08-01 Ethicon, Inc. In vitro expansion of postpartum derived cells in roller bottles
JP2009520466A (en) 2005-12-22 2009-05-28 エニス,ジエーン Viable cells from frozen umbilical cord tissue
EP1979050B1 (en) 2005-12-28 2017-04-19 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using postpartum-derived cells
EP1974013A2 (en) 2005-12-29 2008-10-01 Anthrogenesis Corporation Improved composition for collecting and preserving placental stem cells and methods of using the composition
PL2471904T3 (en) 2005-12-29 2019-08-30 Celularity, Inc. Placental stem cell populations
US8455250B2 (en) 2005-12-29 2013-06-04 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
US20070253931A1 (en) 2006-01-12 2007-11-01 Osiris Therapeutics, Inc. Use of mesenchymal stem cells for treating genetic diseases and disorders
US9944900B2 (en) 2006-01-18 2018-04-17 Hemacell Perfusion Pulsatile perfusion extraction method for non-embryonic pluripotent stem cells
WO2007087293A2 (en) 2006-01-23 2007-08-02 Athersys, Inc. Mapc therapeutics without adjunctive immunosuppressive treatment
ZA200810412B (en) 2006-06-09 2010-03-31 Anthrogenesis Corp Placental niche and use thereof to culture stem cells
US20070287176A1 (en) 2006-06-13 2007-12-13 Alireza Rezania Chorionic villus derived cells
US7993918B2 (en) 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
WO2008021391A1 (en) 2006-08-15 2008-02-21 Anthrogenesis Corporation Umbilical cord biomaterial for medical use
US8122763B2 (en) 2006-09-01 2012-02-28 Avair, Llc Breathing gas supply visual broadcast apparatus
WO2008042441A1 (en) 2006-10-03 2008-04-10 Anthrogenesis Corporation Use of umbilical cord biomaterial for ocular surgery
WO2008060377A2 (en) 2006-10-04 2008-05-22 Anthrogenesis Corporation Placental or umbilical cord tissue compositions
CA3178363A1 (en) 2006-10-06 2008-05-15 Celularity Inc. Human placental collagen compositions, and methods of making and using the same
CA2850793A1 (en) 2006-10-23 2008-05-02 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
CN101611139B (en) 2006-11-13 2012-07-04 伊西康公司 In vitro expansion of postpartum-derived cells using microcarriers
EP2129775A1 (en) 2007-02-12 2009-12-09 Anthrogenesis Corporation Hepatocytes and chondrocytes from adherent placental stem cells; and cd34+, cd45- placental stem cell-enriched cell populations
AU2008216749B2 (en) 2007-02-12 2014-03-13 Celularity Inc. Treatment of inflammatory diseases using placental stem cells
DK2142642T3 (en) 2007-03-27 2017-09-11 Ipd-Therapeutics B V METHODS AND AGENTS FOR STEM CELL PROLIFERATION AND SUBSEQUENT GENERATION AND EXPANSION OF PROGENITOR CELLS, AS WELL AS PRODUCTION OF EFFECT CELLS AS CLINICAL THERAPEUTICS
US20100172830A1 (en) 2007-03-29 2010-07-08 Cellx Inc. Extraembryonic Tissue cells and method of use thereof
KR100902340B1 (en) * 2007-08-02 2009-06-12 한국생명공학연구원 An agent for differentiating hematopoietic stem cell into natural killer cell comprising yc-1 or il-21 and a method of differentiating hematopoietic stem cell into natural killer cell using thereof
KR20220122774A (en) 2007-09-26 2022-09-02 셀룰래리티 인코포레이티드 Angiogenic cells from human placental perfusate
EP2783692B1 (en) 2007-09-28 2019-01-02 Celularity, Inc. Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
MX2010005018A (en) 2007-11-07 2010-05-27 Anthrogenesis Corp Treatment of premature birth complications.
US20090123442A1 (en) 2007-11-09 2009-05-14 Avaris Ab Expanded nk cells
EP2235162B1 (en) * 2008-01-08 2014-10-15 The University Of Queensland Method of producing a population of cells
KR101133185B1 (en) 2008-07-29 2012-04-06 서울대학교병원 Method for Proliferating Natural Killer cell
MX2011001990A (en) 2008-08-20 2011-03-29 Anthrogenesis Corp Improved cell composition and methods of making the same.
CN105796602A (en) 2008-08-20 2016-07-27 人类起源公司 Treatment of stroke using isolated placental cells
CN102176919A (en) 2008-08-22 2011-09-07 人类起源公司 Methods and compositions for treatment of bone defects with placental cell populations
EP2336303B1 (en) * 2008-09-08 2015-07-15 Riken NKT CELL-DERIVED iPS CELLS AND NKT CELLS DERIVED THEREFROM
CN101684456A (en) * 2008-09-28 2010-03-31 江门罗森生物制药有限公司 Method for amplifying NK cells of human beings under condition of in vitro culture
KR101077912B1 (en) * 2008-10-24 2011-10-31 주식회사 메디셀 A method for effective expansion and differentiation of NK cells from Cord Blood
KR20110086176A (en) 2008-11-19 2011-07-27 안트로제네시스 코포레이션 Amnion derived adherent cells
MX2011005230A (en) 2008-11-21 2011-06-16 Anthrogenesis Corp Treatment of diseases, disorders or conditions of the lung using placental cells.
EP2398897B1 (en) * 2009-02-20 2017-06-28 Cellular Dynamics International, Inc. Methods and compositions for the differentiation of stem cells
NZ595440A (en) 2009-03-25 2014-05-30 Anthrogenesis Corp Tumor suppression using human placenta-derived intermediate natural killer cells and immunomodulatory compounds
JP2012521215A (en) 2009-03-26 2012-09-13 アヴァリス・アクチエボラーグ Proliferation of NK cells
US20100323446A1 (en) 2009-06-05 2010-12-23 Jill Renee Barnett Method of collecting placental cells
CA2767014C (en) 2009-07-02 2022-01-25 Anthrogenesis Corporation Method of producing erythrocytes without feeder cells
ES2646750T3 (en) 2010-01-26 2017-12-15 Anthrogenesis Corporation Treatment of bone-related cancers using placental stem cells
US20110280849A1 (en) 2010-03-26 2011-11-17 Anthrogenesis Corporation Tumor suppression using human placenta-derived intermediate natural killer cells and immunomodulatory compounds
SI2556145T1 (en) 2010-04-07 2017-01-31 Anthrogenesis Corporation Angiogenesis using placental stem cells
KR20130092394A (en) 2010-04-08 2013-08-20 안트로제네시스 코포레이션 Treatment of sarcoidosis using placental stem cells
NZ605505A (en) 2010-07-13 2015-02-27 Anthrogenesis Corp Methods of generating natural killer cells
WO2012075412A1 (en) 2010-12-03 2012-06-07 Michael Verneris Generation of natural killer cells and lymphoid tissue inducer-like (lti-like) nk-22 cells
AU2011342904B2 (en) 2010-12-17 2016-05-12 Biolamina Ab Cell culture medium
US20120171295A1 (en) 2010-12-30 2012-07-05 Sascha Abramson Methods for cryopreserving and encapsulating cells
US20120171180A1 (en) 2010-12-30 2012-07-05 Sascha Abramson Compositions comprising amnion derived adherent cells and platelet-rich plasma
WO2012092458A2 (en) 2010-12-30 2012-07-05 Anthrogenesis Corporation Compositions comprising placental stem cells and platelet rich plasma, and methods of use thereof
AR093183A1 (en) 2010-12-31 2015-05-27 Anthrogenesis Corp INCREASE IN THE POWER OF PLACENTA MOTHER CELLS USING MODULATING RNA MOLECULES
EP3443968A1 (en) 2011-06-01 2019-02-20 Celularity, Inc. Treatment of pain using placental stem cells

Also Published As

Publication number Publication date
EP3358007A1 (en) 2018-08-08
KR20210063457A (en) 2021-06-01
JP2020096607A (en) 2020-06-25
WO2012009422A1 (en) 2012-01-19
JP5996533B2 (en) 2016-09-21
KR20130093091A (en) 2013-08-21
KR20180099915A (en) 2018-09-05
US20220259563A1 (en) 2022-08-18
NZ703888A (en) 2016-12-23
CN103097520A (en) 2013-05-08
KR20190108599A (en) 2019-09-24
BR112013000822B1 (en) 2021-02-23
US8926964B2 (en) 2015-01-06
BR112013000822A2 (en) 2016-05-17
CN107760648A (en) 2018-03-06
US9464274B2 (en) 2016-10-11
US20150072425A1 (en) 2015-03-12
KR20220053695A (en) 2022-04-29
CN103097520B (en) 2017-12-05
IL264271A (en) 2019-02-28
JP2013532469A (en) 2013-08-19
JP2017006129A (en) 2017-01-12
CA2804750C (en) 2022-05-10
NZ605505A (en) 2015-02-27
JP2018099122A (en) 2018-06-28
AU2011279201A1 (en) 2013-01-24
KR20230096132A (en) 2023-06-29
IL224204B (en) 2019-02-28
CA2804750A1 (en) 2012-01-19
CN107828727A (en) 2018-03-23
EP2593542B1 (en) 2018-01-03
AU2011279201B2 (en) 2016-01-21
JP2022078099A (en) 2022-05-24
KR20200077613A (en) 2020-06-30
US20170002322A1 (en) 2017-01-05
ES2666746T3 (en) 2018-05-07
MX342995B (en) 2016-10-21
US20190330592A1 (en) 2019-10-31
EP2593542A1 (en) 2013-05-22
US20120148553A1 (en) 2012-06-14

Similar Documents

Publication Publication Date Title
US20220259563A1 (en) Methods of generating natural killer cells
AU2022200976A1 (en) Natural killer cells and uses thereof
AU2021240151A1 (en) Methods of generating natural killer cells

Legal Events

Date Code Title Description
FG Grant or registration